CN110317213B - Diaryl macrocycles as modulators of protein kinases - Google Patents

Diaryl macrocycles as modulators of protein kinases Download PDF

Info

Publication number
CN110317213B
CN110317213B CN201910549014.1A CN201910549014A CN110317213B CN 110317213 B CN110317213 B CN 110317213B CN 201910549014 A CN201910549014 A CN 201910549014A CN 110317213 B CN110317213 B CN 110317213B
Authority
CN
China
Prior art keywords
fluoro
mixture
alkyl
methyl
pyrazolo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201910549014.1A
Other languages
Chinese (zh)
Other versions
CN110317213A (en
Inventor
崔景荣
李一山
埃文·W·罗杰斯
翟大勇
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tepu Pharmaceutical
Original Assignee
Tepu Pharmaceutical
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tepu Pharmaceutical filed Critical Tepu Pharmaceutical
Publication of CN110317213A publication Critical patent/CN110317213A/en
Application granted granted Critical
Publication of CN110317213B publication Critical patent/CN110317213B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Abstract

The present invention relates to diaryl macrocycles as modulators of protein kinases. The present invention relates to certain diaryl macrocycle compounds, pharmaceutical compositions containing them and methods of using them, including methods of treating cancer, pain, neurological diseases, autoimmune diseases and inflammation.

Description

Diaryl macrocycles as modulators of protein kinases
This application is a divisional application of an invention patent application having an application date of 2015, 1/23, application number of 201580005705.5 and an invention name of "diaryl macrocycle as a modulator of protein kinase".
CROSS-REFERENCE TO RELATED APPLICATIONS
The present application claims priority from U.S. provisional application No. 61/931,506, filed on 2014, 24, 2014, 62/049,326, filed on 2014,9, 11, and 62/106,301, filed on 2015, 1, 22, according to 35u.s.c. 119 (e), the entire contents of which are incorporated herein by reference in their entirety.
Technical Field
The present invention relates to certain diaryl macrocyclic derivatives, pharmaceutical compositions containing them and their use in the treatment of cancer, pain, neurological diseases, autoimmune diseases and inflammation.
Background
Protein kinases are key regulators of cell growth, proliferation and survival. Genetic and epigenetic changes accumulate in cancer cells, which result in abnormal activation of signal transduction pathways that drive malignant processes. Manning G. (Manning, G) et al, science 2002,298,1912-1934. Pharmacological inhibition of these signaling pathways represents a promising opportunity for intervention in targeted cancer therapies. Soyes c. (Sawyers, c.), nature (Nature) 2004,432,294-297.
MET as well as RON belong to a unique subset of receptor tyrosine kinases and are produced primarily in cells of epithelial or endothelial origin. Park M. (Park, M.) et al, cell (Cell) 1986,45,895-904. Hepatocyte Growth Factor (HGF), also known as Scatter Factor (SF), is the only known natural high affinity ligand for MET and is expressed predominantly in cells of mesenchymal origin. Botaro d.p. (Bottaro, d.p.) et al, science 1991,251,802-804.HGF/MET signaling controls MET-dependent cell proliferation, survival and migration processes that are critical for invasive growth during embryonic development and postnatal organ regeneration, and are fully activated in adults only for wound healing and tissue regeneration processes. Tasolino l. (Trusolino, l.) et al, review by Nature-molecular cell biology (Nature rev. Mol. Cell biol.) -2010, 11,834-848. The HGF/MET axis is often upregulated in many cancers by activating mutations, gene amplification, aberrant paracrine or autocrine ligand production, and is strongly associated with tumorigenesis, invasive growth and metastasis. Galradi E. (Gherardi, E.) et al, nature review-cancer (Nature Rev. Cancer) 2012,12,89-103. Furthermore, activation of HGF/MET signaling is becoming an important mechanism for treatment against EGFR and BRAF inhibitors via MET amplification and/or upregulation of matrix HGF. Engelman j.a. (Engelman, j.a.), et al, science 2007,316,1039-1043; wilson, t.r. et al, 2012,487,505-509. Due to the role of aberrant HGF/MET signaling in human tumorigenesis, invasion/metastasis and resistance, inhibition of the HGF/MET signaling pathway has great potential in cancer therapy.
ALK and Leukocyte Tyrosine Kinases (LTK) are grouped within the Insulin Receptor (IR) superfamily of receptor tyrosine kinases. ALK is predominantly expressed in the central and peripheral nervous systems, suggesting a potential role in the normal development and function of the nervous system. Pulford K. (Pulford, K.) et al, cell and molecular Life sciences (Cell mol. Life Sci.) 2004,61,2939.ALK was first discovered as a fusion protein NPM (nucleolar phosphoprotein) -ALK encoded by a fusion gene resulting from a t (2. Morris S.W. (Morris, S.W.) et al, science 1994,263,1281. More than twenty different ALK translocation partners have been found in many cancers, including ALCL (60-90% incidence), inflammatory myofibroblast tumors (IMT, 50-60%), non-small cell lung cancer (NSCLC, 3-7%), colorectal cancer (CRC, 0-2.4%), breast cancer (0-2.4%) and others. Grande e et al, molecular cancer therapeutics (mol. Cancer ther.) 2011,10,569-579. ALK-fusion proteins are located in the cytoplasm, and fusion partners with ALK function in dimerization or oligomerization of the fusion protein via helix-helix interactions to generate constitutive activation of ALK kinase function. Bischof D.et al, molecular cell biology, 1997,17,2312-2325.EML4-ALK, which contains portions of the echinoderm microtubule-associated protein-like 4 (EML 4) gene and the ALK gene, was first found in NSCLC, is highly oncogenic, and was shown to cause lung adenocarcinoma in transgenic mice. Soda M. (Soda, M.) et al, nature 2007,448,561-566. Oncogenic point mutations of ALK in both familial and sporadic cases of neuroblastoma. Moss Y.P. (Moss, Y.P.), et al, nature 2008,455,930-935. ALK is an attractive molecular target for cancer therapeutic intervention due to its important role in hematopoietic, solid, and interstitial tumors. And gelland, supra.
Tropomyosin-related receptor tyrosine kinase (Trk) is a high affinity receptor for Neurotrophin (NT), a Nerve Growth Factor (NGF) family of proteins. Members of the Trk family are highly expressed in cells of neural origin. Trk (TrkA, trkB, and TrkC) mediates the survival and differentiation of neurons during development through its activation of preferably neurotrophins (NGF against TrkA, brain-derived neurotrophic factors [ BDNF ] and NT4/5 against TrkB, and NT3 against TrkC). The NT/Trk signaling pathway functions to protect the endogenous system of neurons following biochemical damage, transient ischemia, or physical injury. Thele c.j. (Thiele, c.j.) et al, clinical cancer research (clin. Cancer res.) 2009,15,5962-5967. However, trk was originally cloned in the extracellular domain as an oncogene fused with the tropomyosin gene. Activating mutations caused by chromosomal rearrangements or mutations in NTRK1 (TrkA) have been identified in papillary and myelothyroid carcinomas, and recently in non-small cell lung cancer. Picodi m.a. (Pierotti, m.a.) et al, cancer bulletin (Cancer lett.) 2006,232,90-98; waschnawei A. (Vaishnavi, A.), et al, nature medicine (nat. Med.) 2013,19,1469-1472. Since Trk plays an important role in pain perception as well as tumor cell growth and survival signaling, inhibitors of Trk receptor kinases may provide benefits as therapeutic agents for pain and cancer.
The receptor tyrosine kinase AXL belongs to the TAM family of proteins and was originally detected as an unidentified transformed gene in patients with Chronic Myelogenous Leukemia (CML). Verma, a. (Verma, a.), et al, molecular cancer therapeutics 2011,10,1763-1773. The primary ligand for the TAM receptor is the growth arrest specific 6 protein (Gas 6). AXL is widely expressed and has been detected in a variety of organs and cells, including hippocampus and cerebellum, monocytes, macrophages, platelets, endothelial Cells (EC), heart, skeletal muscle, liver, kidney, and testis. Upregulation of Gas6/AXL has been reported in many human cancers, including colon, esophagus, thyroid, breast, lung, liver and astrocytoma-malignant glioma pleomorphis. The references are as above. Increased activation of AXL and acquired resistance to erlotinib (erlotinib) in the absence of EGFR T790M change or MET activation have been observed in vitro and in vivo in EGFR-mutant lung cancer models. Zhang Z. (Zhang, Z.) et al, nature genetics (nat. Genet.) 2012,44,852-860. In these tumor models, genetic or pharmacological inhibition of AXL restores sensitivity to erlotinib. Increased expression of AXL and in some cases its ligand Gas6 was found in EGFR mutant lung cancers obtained from individuals with acquired resistance to tyrosine kinase inhibitors. The references are as above. AXL is therefore a promising therapeutic target for EGFR mutant lung cancer patients to gain resistance to EGFR inhibitors.
Crizotinib (Crizotinib) (PF-02341066) is a MET/ALK/ROS1/RON targeted tyrosine kinase drug with moderate activity against TRK and AXL. J.J. (Cui, J.J.), et al, J.Med.chem., 2011,54,6342-6363. It has been approved for the treatment of certain patients with advanced (locally advanced or metastatic) NSCLC expressing an abnormal ALK fusion gene identified by a companion diagnostic test (the vises ALK Break Apart FISH Probe Kit). Similar to imatinib (imatinib) and other kinase inhibitor drugs, resistance invariably develops after a certain period of treatment with crizotinib. Resistance mechanisms include ALK gene amplification, secondary ALK mutations, and abnormal activation of other kinases including KIT and EGFR. Shishan r. (Katayama, r.) et al, 2012,4,120ra17, scientific translation medicine (sci. Second-generation ALK inhibitors are emerging based on the clinical success of second-generation ABL inhibitors for the treatment of imatinib resistance in CML patients. These drugs are targeted at treating patients with refractory or resistant NSCLC to crizotinib, with more effective inhibition of both wild and mutant ALK proteins. Gridelli c. (Gridelli, c.) et al, cancer therapy review (Cancer Treat rev.) 2014,40,300-306.
By modulating multiple targets in a group of structurally related tyrosine kinases MET, ALK, AXL and TRK, the compounds described herein address crizotinib resistance, EGFR inhibitor drug resistance, and other major indications of having aberrant cell signaling due to MET, ALK, AXL and/or TRK mutations and gene amplification. The compounds described herein are inhibitors of MET, wild and mutant ALK, AXL and TRK and would be useful in treating cancer patients with aberrant signaling from one or more of MET, ALK, AXL or TRK.
The Janus kinase family (JAK) includes JAK1, JAK2, JAK3 and TYK2, and are cytoplasmic tyrosine kinases required for physiological signaling of cytokines and growth factors. Quintas-cadama, et al (Quintas-Cardama, a.), reviews by nature: drug discovery (nat. Rev. Drug discov.) 2011,10 (2), 127-40; pesu m. (Pesu, m.) et al, immunological reviews (immunol. Rev.) -2008, 223,132-142; murray p.j. (Murray, p.j.), journal of immunology (j.immunol.) 2007,178 (5), 2623-2329. JAKs are activated by ligand-induced oligomerization, which results in activation of downstream transcriptional signaling pathways called STATs (signal transducers and activators of transcription)). Phosphorylated STATs dimerize and translocate into the nucleus to drive the expression of specific genes involved in proliferation, apoptosis, differentiation, which are essential for hematopoiesis, inflammation, and immune responses. Merry, see above.
Mouse gene knockout studies have implicated a major role for JAK-STAT signaling, with some overlap between them. JAK1 plays a key role in the signaling of various proinflammatory cytokines (e.g., IL-1, IL-4, IL-6) and tumor necrosis factor alpha (TNF α). Muller M. (Muller, M.) et al, nature 1993,366 (6451), 129-135.JAK2 is used for hematopoietic growth factor signaling, such as Epo, IL-3, IL-5, GM-CSF, thrombopoietin, growth hormone, and prolactin-mediated signaling. Nouy Baer H. (Neubauer, H.), et al, cell 1998 93 (3), 397-409.JAK3 plays a role in mediating immune responses, and TYK2 associates with JAK2 or JAK3 to transduce signals of cytokines (e.g., IL-12). Yeban T. (Nosaka, T.), et al, science 1995,270 (5237), 800-802; wainschen, w. (Vainchenker, w.) et al, seminal institute of cell developmental biology (semin. 2008,19 (4),385-393.
Aberrant regulation of the JAK/STAT pathway has been implicated in a variety of human pathological diseases, including cancer (JAK 2) and rheumatoid arthritis (JAK 1, JAK 3). A functionally acquired mutation of JAK2 (JAK 2V 617F) has been observed at high frequency in MPN patients. Levin r.l. (Levine, r.l.) et al, cancer cells (Cancer cells) 2005,7 (4), 387-397; kralovas r. (Kralovics, r.), et al, new england journal of medicine (n.engl.j.med.) 2005,253 (17), 1779-1790; james c. (James, c.) et al, nature 2005,434 (7037), 1144-1148; baxter, E.J., et al, lancet 2005,365 (9464), 1054-1061. Mutations in the JH2 pseudo-kinase domain of JAK2 result in constitutive kinase activity. Cells containing the JAK2V617F mutation acquire cytokine independent growth capacity and often become tumors, which provides a powerful basis for developing JAK inhibitors as target therapies.
Many JAK inhibitors have shown significant benefits in clinical trials for myelofibrotic patients in splenomegaly and disease-related systemic symptoms, including the first JAK2 inhibitor ruxolitinib approved by the FDA in 2011. Kuntas-kammy, supra; sobol m.b. (Sonbol, m.b.) et al, hematology research progress (ther. Adv.hematol.) 2013,4 (1), 15-35; latawin l.m. (LaFave, l.m.), et al, trends in pharmaceutical sciences (Trends pharmacol. Sci.) 2012,33 (11), 574-582. Recently collected clinical data on ruxolitinib treatment indicated that JAK inhibitors act in both JAK2 wild-type and JAK2 mutated situations. Verstovsek s. (Verstovsek, s.) et al, new england journal of medicine 2012,366 (9), 799-807; kunststo-Karma motor A. Et al, blood (Blood) 2010,115 (15), 3109-3117. The discovery of selective inhibitors of JAK2 to JAK1/3 remains an unresolved challenge. In addition, hyperactivation of JAK 2/signal transduction and activator of transcription 3 (JAK 2/STAT 3) is responsible for abnormal dendritic cell differentiation, which is a condition that results in abnormal dendritic cell differentiation and the accumulation of immunosuppressive myeloid cells in cancer (Nefedova Y., et al, cancer research 2005 (20): 9525-35). In Pten-null aging tumors, activation of the Jak2/Stat3 pathway establishes an immunosuppressive tumor microenvironment that contributes to tumor growth and chemoresistance (Toso, a.). Et al, cell Reports 2014,9, 75-89. Therefore, pharmacological inhibition of the JAK2/STAT3 pathway may be an important new therapeutic strategy to enhance antitumor activity via modulation of antitumor immunity.
ROS1 kinase is a receptor tyrosine kinase with unknown ligands. The normal function of human ROS1 kinase is not fully understood. However, ROS1 kinase has been reported to undergo gene rearrangement to produce constitutively active fusion proteins in a number of human cancers including glioblastoma, non-small cell lung cancer (NSCLC), cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumors, angiosarcoma, and epithelioid vascular endothelial tumors (Davies, k.d., et al, clinical cancer studies 2013,19 (15): 4040-4045). Targeting ROS1 fusion proteins with crizotinib has demonstrated promising clinical efficacy in NSCLC patients whose tumors are positive for ROS1 genetic abnormalities (Shaw, a.t.) et al, new england journal of medicine 2014,371 (21): 1963-1971). Acquired resistance mutations have been observed in crizotinib-treated patients (adrad m.m. (Awad, m.m.) et al, new england journal of medicine 2013,368 (25): 2396-2401). There is an urgent need to develop second generation ROS1 inhibitors for overcoming crizotinib ROS1 resistance.
There remains a need for small molecule inhibitors of these various protein or tyrosine kinase targets with desirable pharmaceutical properties. In the context of the present invention, it has been found that certain diaryl macrocycle compounds have this advantageous activity property.
Disclosure of Invention
In one aspect, the invention relates to compounds having the following formula (I-A):
Figure GDA0003692253770000051
wherein
Ring a 'and ring B' are each independently monocyclic or bicyclic aryl or heteroaryl; wherein one of ring a 'and ring B' is monocyclic aryl or heteroaryl and the other is bicyclic heteroaryl; and at least one of ring a 'and ring B' comprises at least one nitrogen ring member;
each L 1 And L 2 Independently is-C (R) 1’ )(R 2’ )-、-O-、-N(R k’ ) -, -S-, -S (O) -or-S (O) 2 -;
Each R 1’ And R 2’ Independently of each other H, deuterium, halogen, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl OR monocyclic OR bicyclic heteroaryl, -OR a’ 、-OC(O)R a’ 、-OC(O)NR a’ R b’ 、-OS(O)R a’ 、-OS(O) 2 R a’ 、-SR a’ 、-S(O)R a’ 、-S(O) 2 R a’ 、-S(O)NR a’ R b’ 、-S(O) 2 NR a’ R b’ 、-OS(O)NR a’ R b’ 、-OS(O) 2 NR a’ R b’ 、-NR a’ R b’ 、-NR a’ C(O)R b’ 、-NR a’ C(O)OR b’ 、-NR a’ C(O)NR a’ R b’ 、-NR a’ S(O)R b’ 、-NR a’ S(O) 2 R b’ 、-NR a’ S(O)NR a’ R b’ 、-NR a’ S(O) 2 NR a’ R b’ 、-C(O)R a’ 、-C(O)OR a’ 、-C(O)NR a’ R b’ 、-PR a’ R b’ 、-P(O)R a’ R b’ 、-P(O) 2 R a’ R b’ 、-P(O)NR a’ R b’ 、-P(O) 2 NR a’ R b’ 、-P(O)OR a’ 、-P(O) 2 OR a’ -CN or-NO 2 Or R is 1’ And R 2’ Together with the carbon or carbons to which they are attached form C 3-6 Cycloalkyl or 4-to 6-membered heterocycloalkyl, wherein C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in aryl, monocyclic or bicyclic heteroaryl, 4-to 6-membered heterocycloalkyl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR e’ 、-OC(O)R e’ 、-OC(O)NR e’ R f’ 、-OS(O)R e’ 、-OS(O) 2 R e’ 、-OS(O)NR e’ R f’ 、-OS(O) 2 NR e’ R f’ 、-SR e’ 、-S(O)R e’ 、-S(O) 2 R e’ 、-S(O)NR e’ R f’ 、-S(O) 2 NR e’ R f’ 、-NR e’ R f’ 、-NR e’ C(O)R f’ 、-NR e’ C(O)OR f’ 、-NR e’ C(O)NR e’ R f’ 、-NR e’ S(O)R f’ 、-NR e’ S(O) 2 R f’ 、-NR e’ S(O)NR e’ R f’ 、-NR e’ S(O) 2 NR e’ R f’ 、-C(O)R e’ 、-C(O)OR e’ 、-C(O)NR e’ R f’ 、-PR e’ R f’ 、-P(O)R e’ R f’ 、-P(O) 2 R e’ R f’ 、-P(O)NR e’ R f’ 、-P(O) 2 NR e’ R f’ 、-P(O)OR e’ 、-P(O) 2 OR e’ -CN or-NO 2
Each R k’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl, wherein C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in the aryl or monocyclic or bicyclic heteroaryl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR e’ 、-OC(O)R e’ 、-OC(O)NR e’ R f’ 、-OS(O)R e’ 、-OS(O) 2 R e’ 、-OS(O)NR e’ R f’ 、-OS(O) 2 NR e’ R f’ 、-SR e’ 、-S(O)R e’ 、-S(O) 2 R e’ 、-S(O)NR e’ R f’ 、-S(O) 2 NR e’ R f’ 、-NR e’ R f’ 、-NR e’ C(O)R f’ 、-NR e’ C(O)OR f’ 、-NR e’ C(O)NR e’ R f’ 、-NR e’ S(O)R f’ 、-NR e’ S(O) 2 R f’ 、-NR e’ S(O)NR e’ R f’ 、-NR e’ S(O) 2 NR e’ R f’ 、-C(O)R e’ 、-C(O)OR e’ 、-C(O)NR e’ R f’ 、-PR e’ R f’ 、-P(O)R e’ R f’ 、-P(O) 2 R e’ R f’ 、-P(O)NR e’ R f’ 、-P(O) 2 NR e’ R f’ 、-P(O)OR e’ 、-P(O) 2 OR e’ -CN or-NO 2
Each R 3’ And R 4’ Independently of each other, is deuterium, halogen, -OR c’ 、-OC(O)R c’ 、-OC(O)NR c’ R d’ 、-OC(=N)NR c’ R d’ 、-OS(O)R c’ 、-OS(O) 2 R c’ 、-OS(O)NR c’ R d’ 、-OS(O) 2 NR c’ R d’ 、-SR c’ 、-S(O)R c’ 、-S(O) 2 R c’ 、-S(O)NR c’ R d’ 、-S(O) 2 NR c’ R d’ 、-NR c’ R d’ 、-NR c’ C(O)R d’ 、-NR c’ C(O)OR d’ 、-NR c’ C(O)NR c’ R d’ 、-NR c’ C(=N)NR c’ R d’ 、-NR c’ S(O)R d’ 、-NR c’ S(O) 2 R d’ 、-NR c’ S(O)NR c’ R d’ 、-NR c’ S(O) 2 NR c’ R d’ 、-C(O)R c’ 、-C(O)OR c’ 、-C(O)NR c’ R d’ 、-C(=N)NR c’ R d’ 、-PR c’ R d’ 、-P(O)R c’ R d’ 、-P(O) 2 R c’ R d’ 、-P(O)NR c’ R d’ 、-P(O) 2 NR c’ R d’ 、-P(O)OR c’ 、-P(O) 2 OR c’ 、-CN、-NO 2 、C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl, or any two R 3’ A group or any two of R 4’ The group together with the ring to which it is attached forms C 5-8 Cycloalkyl or 5-to 8-membered heterocycloalkyl, wherein C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl, monocyclic or bicyclic heteroaryl C 5-8 Each hydrogen atom in cycloalkyl or 5-to 8-membered heterocycloalkyl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR e’ 、-OC(O)R e’ 、-OC(O)NR e’ R f’ 、-OS(O)R e’ 、-OS(O) 2 R e’ 、-OS(O)NR e’ R f’ 、-OS(O) 2 NR e’ R f’ 、-SR e’ 、-S(O)R e’ 、-S(O) 2 R e’ 、-S(O)NR e’ R f’ 、-S(O) 2 NR e’ R f’ 、-NR e’ R f’ 、-NR e’ C(O)R f’ 、-NR e’ C(O)OR f’ 、-NR e’ C(O)NR e’ R f’ 、-NR e’ S(O)R f’ 、-NR e’ S(O) 2 R f’ 、-NR e’ S(O)NR e’ R f’ 、-NR e’ S(O) 2 NR e’ R f’ 、-C(O)R e’ 、-C(O)OR e’ 、-C(O)NR e’ R f’ 、-PR e’ R f’ 、-P(O)R e’ R f’ 、-P(O) 2 R e’ R f’ 、-P(O)NR e’ R f’ 、-P(O) 2 NR e’ R f’ 、-P(O)OR e’ 、-P(O) 2 OR e’ -CN or-NO 2
R 7’ Is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl, wherein C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in the aryl or monocyclic or bicyclic heteroaryl is independently optionally substituted by: deuterium, halogen, -OR i’ 、-OC(O)R i’ 、-OC(O)NR i’ R j’ 、-OS(O)R i’ 、-OS(O) 2 R i’ 、-OS(O)NR i’ R j’ 、-OS(O) 2 NR i’ R j’ 、-SR i’ 、-S(O)R i’ 、-S(O) 2 R i’ 、-S(O)NR i’ R j’ 、-S(O) 2 NR i’ R j’ 、-NR i’ R j’ 、-NR i’ C(O)R j’ 、-NR i’ C(O)OR j’ 、-NR i’ C(O)NR i’ R j’ 、-NR i’ S(O)R j’ 、-NR i’ S(O) 2 R j’ 、-NR i’ S(O)NR i’ R j’ 、-NR i’ S(O) 2 NR i’ R j’ 、-C(O)R i’ 、-C(O)OR i’ 、-C(O)NR i’ R j’ 、-PR i’ R j’ 、-P(O)R i’ R j’ 、-P(O) 2 R i’ R j’ 、-P(O)NR i’ R j’ 、-P(O) 2 NR i’ R j’ 、-P(O)OR i’ 、-P(O) 2 OR i’ CN or NO 2
Each R a’ 、R b’ 、R c’ 、R d’ 、R e’ 、R f’ 、R i’ And R j’ Independently selected from the group consisting of: H. deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl and heteroaryl;
m' is 2,3, 4 or 5;
n' is 2,3 or 4;
p' is 0,1, 2,3 or 4; and is
q' is 0,1, 2,3 or 4;
or a pharmaceutically acceptable salt thereof.
In one aspect, the invention relates to a chemical entity of formula (I-A):
Figure GDA0003692253770000081
wherein
Ring a 'and ring B' are each independently monocyclic or bicyclic aryl or heteroaryl;
wherein one of ring a 'and ring B' is monocyclic aryl or heteroaryl and the other is bicyclic heteroaryl; and at least one of ring a 'and ring B' comprises at least one nitrogen ring member;
each R 3’ And R 4’ Independently of each other, is deuterium, halogen, -OR c’ 、-OC(O)R c’ 、-OC(O)NR c’ R d’ 、-OC(=N)NR c’ R d’ 、-OS(O) 0-2 R c’ 、-OS(O) 0-2 NR c’ R d’ 、-S(O) 0-2 R c’ 、-S(O) 0-2 NR c’ R d’ 、-NR c’ R d’ 、-NR c’ C(O)R d’ 、-NR c’ C(O)NR c’ R d’ 、-NR c’ C(=N)NR c’ R d’ 、-NR c’ S(O) 0-2 R d’ 、-NR c’ S(O) 0-2 NR c’ R d’ 、-C(O)R c’ 、-C(O)OR c’ 、-C(O)NR c’ R d’ 、-C(=N)NR c’ R d’ 、-P(O) 0-2 R c’ R d’ 、-P(O) 0-2 NR c’ R d’ 、-P(O) 0-2 OR c’ 、-CN、-NO 2 、C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl; or any two of R 3’ A group or any two of R 4’ The group together with the ring to which it is attached forms C 5-8 Cycloalkyl or 5 to 8 membered heterocycloalkyl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, and mono-or bicyclic heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR e’ 、-OC(O)R e’ 、-OC(O)NR e’ R f’ 、-OS(O) 0-2 R e’ 、-OS(O) 0-2 NR e’ R f’ 、-S(O) 0-2 R e’ 、-S(O) 0-2 NR e’ R f’ 、-NR e’ R f’ 、-NR e’ C(O)R f’ 、-NR e’ C(O)NR e’ R f’ 、-NR e’ S(O) 0-2 R f’ 、-NR e’ S(O) 0-2 NR e’ R f’ 、-C(O)R e’ 、-C(O)OR e’ 、-C(O)NR e’ R f’ 、-P(O) 0-2 R e’ R f’ 、-P(O) 0-2 NR e’ R f’ 、-P(O) 0-2 OR e’ CN and NO 2 (ii) a And is
Each R c’ 、R d’ 、R e’ And R f’ Independently selected from the group consisting of: H. deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl and heteroaryl;
R 7’ is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, or heteroaryl is substituted or unsubstituted with one or more substituents selected from the group consisting of: deuterium, halogen, -OR i’ 、-OC(O)R i’ 、-OC(O)NR i’ R j’ 、-OS(O) 0-2 R i’ 、-OS(O) 0-2 NR i’ R j’ 、-S(O) 0-2 R i’ 、-S(O) 0-2 NR i’ R j’ 、-NR i’ R j’ 、-NR i’ C(O)R j’ 、-NR i’ C(O)NR i’ R j’ 、-NR i’ S(O) 0-2 R j’ 、-NR i’ S(O) 0-2 NR i’ R j’ 、-C(O)R i’ 、-C(O)OR i’ 、-C(O)NR i’ R j’ 、-P(O) 0-2 R i’ R j’ 、-P(O) 0-2 NR i’ R j’ 、-P(O) 0-2 OR i’ -CN and-NO 2
Wherein each R is i’ And R j’ Independently of each other H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl;
each L 1 And L 2 Independently is-C (R) 1’ )(R 2’ )-、-O-、-N(R k’ ) -or-S (O) 0-2
Wherein each R is 1’ And R 2’ Independently of each other is H, deuterium, halogen, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl; or R 1’ And R 2’ Together with the carbon or carbons to which they are attached form C 3-6 Cycloalkyl or 4 to 6 membered heterocycloalkyl;
each R k’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl;
wherein R is 1’ 、R 2’ Or R k Each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, or heteroaryl in' is unsubstituted or substituted with one or more substituents selected from the group consisting of: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR a’ 、-OC(O)R a’ 、-OC(O)NR a’ R b’ 、-OS(O) 0-2 R a’ 、-OS(O) 0-2 NR a’ R b’ 、-S(O) 0-2 R a’ 、-S(O) 0- 2 NR a’ R b’ 、-NR a’ R b’ 、-NR a’ C(O)R b’ 、-NR a’ C(O)NR a’ R b’ 、-NR a’ S(O) 0-2 R b’ 、-NR a’ S(O) 0-2 NR a’ R b’ 、-C(O)R a’ 、-C(O)OR a’ 、-C(O)NR a’ R b’ 、-P(O) 0-2 R a’ R b’ 、-P(O) 0-2 NR a’ R b’ 、-P(O) 0-2 OR a’ -CN and-NO 2
Wherein each R is a’ And R b’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or heteroaryl;
m' is 3,4 or 5;
n' is 2,3 or 4;
p' is 0,1, 2,3 or 4; and is
q' is 0,1, 2,3 or 4;
or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to a chemical entity of formula (I):
Figure GDA0003692253770000091
wherein
Ring a and ring B are each independently monocyclic or bicyclic aryl or heteroaryl; wherein one of ring a and ring B is monocyclic and the other is bicyclic; and the ring comprises at least one nitrogen ring member;
R 1 and R 2 Each independently of the other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl; or R 1 And R 2 Together with the carbon to which it is attached form C 3-6 Cycloalkyl or 4 to 6 membered heterocycloalkyl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, or heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR a 、-OC(O)R a 、-OC(O)NR a R b 、-OS(O) 0-2 R a 、-OS(O) 0-2 NR a R b 、-NR a R b 、-NR a C(O)R b 、-NR a C(O)NR a R b 、-NR a S(O) 0-2 R b 、-NR a S(O) 0-2 NR a R b 、-C(O)R a 、-C(O)OR a 、-C(O)NR a R b 、-P(O) 0-2 R a R b 、-P(O) 0-2 NR a R b 、-P(O) 0-2 OR a -CN and-NO 2
Wherein each R is a And R b Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or heteroaryl;
each R 3 And R 4 Independently of each other, is deuterium, halogen, -OR c 、-OC(O)R c 、-OC(O)NR c R d 、-OC(=N)NR c R d 、-OS(O) 0-2 R c 、-OS(O) 0-2 NR c R d 、-NR c R d 、-NR c C(O)R d 、-NR c C(O)NR c R d 、-NR c C(=N)NR c R d 、-NR c S(O) 0- 2 R d 、-NR c S(O) 0-2 NR c R d 、-C(O)R c 、-C(O)OR c 、-C(O)NR c R d 、-C(=N)NR c R d 、-P(O) 0-2 R c R d 、-P(O) 0- 2 NR c R d 、-P(O) 0-2 OR c 、-CN、-NO 2 、C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenylNaphthyl and monocyclic or bicyclic heteroaryl are unsubstituted or substituted with one or more substituents selected from the group consisting of: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR e 、-OC(O)R e 、-OC(O)NR e R f 、-OS(O) 0-2 R e 、-OS(O) 0-2 NR e R f 、-NR e R f 、-NR e C(O)R f 、-NR e C(O)NR e R f 、-NR e S(O) 0-2 R f 、-NR e S(O) 0-2 NR e R f 、-C(O)R e 、-C(O)OR e 、-C(O)NR e R f 、-P(O) 0-2 R e R f 、-P(O) 0-2 NR e R f 、-P(O) 0-2 OR e -CN and-NO 2 (ii) a And is provided with
Each R c 、R d 、R e And R f Independently selected from the group consisting of: H. deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl and heteroaryl;
R 5 and R 6 Each independently of the other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl; or R 5 And R 6 Together with the carbon to which it is attached form C 3-6 Cycloalkyl or 4 to 6 membered heterocycloalkyl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, or heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR g 、-OC(O)R g 、-OC(O)NR g R h 、-OS(O) 0-2 R g 、-OS(O) 0-2 NR g R h 、-NR g R h 、-NR g C(O)R h 、-NR g C(O)NR g R h 、-NR g S(O) 0-2 R h 、-NR g S(O) 0-2 NR g R h 、-C(O)R g 、-C(O)OR g 、-C(O)NR g R h 、-P(O) 0-2 R g R h 、-P(O) 0-2 NR g R h 、-P(O) 0-2 OR g -CN and-NO 2
Wherein each R is g And R h Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl or monocyclic or bicyclic heteroaryl;
R 7 is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl;
wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, phenyl, naphthyl, or heteroaryl is substituted or unsubstituted with one or more substituents selected from the group consisting of: deuterium, halogen, -OR i 、-OC(O)R i 、-OC(O)NR i R j 、-OS(O) 0-2 R i 、-OS(O) 0-2 NR i R j 、-NR i R j 、-NR i C(O)R j 、-NR i C(O)NR i R j 、-NR i S(O) 0-2 R j 、-NR i S(O) 0- 2 NR i R j 、-C(O)R i 、-C(O)OR i 、-C(O)NR i R j 、-P(O) 0-2 R i R j 、-P(O) 0-2 NR i R j 、-P(O) 0-2 OR i -CN and-NO 2
Wherein each R is i And R j Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl;
x and Y are each independently-C (R) k )(R k ) -, -O-or-N (R) k )-;
Wherein each R is k Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, phenyl, naphthyl, or a monocyclic or bicyclic heteroaryl;
m is 2,3 or 4;
n is 1,2 or 3;
p is 0,1, 2,3 or 4; and is
q is 0,1, 2,3 or 4;
or a pharmaceutically acceptable salt thereof.
In certain embodiments, the compound of formula (I) or (I-a) is a compound selected from those described or exemplified in the detailed description below.
In certain embodiments, the compound of formula (I) or (I-A) is a compound having the formula
Figure GDA0003692253770000111
Or a pharmaceutically acceptable salt thereof.
In certain embodiments, the compound of formula (I) or (I-A) is a compound having the formula
Figure GDA0003692253770000112
Or a pharmaceutically acceptable salt thereof
In certain embodiments, the compound of formula (I) or (I-A) is a compound of the formula
Figure GDA0003692253770000121
Or a pharmaceutically acceptable salt thereof.
In certain embodiments, the compound of formula (I) or (I-A) is a compound having the formula
Figure GDA0003692253770000122
Or a pharmaceutically acceptable salt thereof.
In certain embodiments, the compound of formula (I) or (I-A) is a compound having the formula
Figure GDA0003692253770000123
Or a pharmaceutically acceptable salt thereof.
In certain embodiments, the compound of formula (I) or (I-A) is a compound having the formula
Figure GDA0003692253770000124
Or a pharmaceutically acceptable salt thereof.
In other aspects, the invention relates to crystalline forms of the free base of the compound of the formula
Figure GDA0003692253770000125
Which has a powder X-ray diffraction pattern substantially the same as that of figure 1. In some embodiments, crystalline polymorph 1 of a free base of a compound of the formula
Figure GDA0003692253770000131
Wherein the powder X-ray diffraction pattern has a peak at a diffraction angle (2 θ) of 21.94. In some embodiments, polymorph 1 of the free base of a compound of the formula
Figure GDA0003692253770000132
Wherein the powder X-ray diffraction pattern has peaks at diffraction angles (2 θ) of 21.94 and 23.96. In some embodiments, polymorph 1 of the free base of a compound of the formula
Figure GDA0003692253770000133
Wherein the powder X-ray diffraction pattern has peaks at diffraction angles (2 θ) of 21.94, 23.96 and 19.64.
In other aspects, the invention relates to pharmaceutical compositions comprising at least one compound of formula (I) or (I-A), or a pharmaceutically acceptable salt thereof. The pharmaceutical composition of the present invention may further comprise a pharmaceutically acceptable excipient. The invention also relates to compounds of formula (I) or (I-a), or a pharmaceutically acceptable salt thereof, for use as a medicament.
In another aspect, the present invention relates to a method of treating cancer, pain, a neurological disease, an autoimmune disease, or inflammation, comprising administering to a patient in need of such treatment an effective amount of at least one compound of formula (I) or (I-a), or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to the use of compounds of formula (I) or (I-A) for the manufacture of medicaments for the treatment of said diseases and medical conditions and the use of said compounds and salts for the treatment of said diseases and medical conditions.
In another aspect, the invention relates to a method of inhibiting protein or tyrosine kinases including one or more of MET, ALK, ROS1, AXL, TRK and JAK comprising contacting a cell comprising one or more of said kinases with an effective amount of at least one compound of formula (I) or (I-a) or a salt thereof, and/or with at least one pharmaceutical composition of the invention, wherein said contacting is in vitro, ex vivo or in vivo.
Additional embodiments, features and advantages of the invention will be apparent from the detailed description and from the practice of the invention.
The disclosures of the publications (including patents) cited in this specification are hereby incorporated by reference for the sake of brevity.
Drawings
FIG. 1 shows the powder X-ray diffraction pattern of crystalline polymorph 1 of the free base of 11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one (example 20).
FIG. 2 shows a differential scanning calorimetry thermogram of crystalline polymorph 1 of the free base of 11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one (example 20).
Detailed Description
Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents, applications, published applications and other publications mentioned herein are incorporated by reference in their entirety. If a definition set forth in this section is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, or other publications that are incorporated herein by reference, the definition set forth in this section prevails over the definition that is incorporated herein by reference.
As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be designed without the inclusion of any optional elements. Accordingly, this statement, together with the statements of the elements of the claims, is intended to serve as antecedent basis for use of exclusive terminology such as "solely," "solely," and the like, or use of "negative" limitations.
As used herein, the terms "comprising," "including," and "containing" are used in their open, non-limiting sense.
To provide a more concise description, some of the quantitative representations presented herein are not limited to the term "about". It is understood that each quantity given herein is intended to refer to the actual given value, and also to the approximation to be reasonably inferred based on the skilled person of the given value, whether or not the term "about" is explicitly used, including equivalents and approximations due to the experimental and/or measurement conditions for the given value. Whenever a yield is given as a percentage, the yield refers to the mass of the entity giving the yield relative to the maximum amount that can be obtained for the same entity under specific stoichiometric conditions. Unless otherwise indicated, concentrations given in percentages refer to mass ratios.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
Unless otherwise indicated, the methods and techniques of the present invention are generally performed according to conventional methods that are well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, for example, loudon (Loudon), organic Chemistry (Organic Chemistry), 4 th edition, new York: oxford University Press, 2002, pages 360-361, 1084-1085; smith (Smith) and marque (March), the advanced organic chemistry of marque: reactions, mechanisms and structures (March's Advanced Organic Chemistry: reactions, mechanics, and Structure), fifth edition, wiley-Interscience, 2001.
Chemical nomenclature for the compounds described herein generally results from the use of commercially available ACD/Name 2014 (ACD/Labs) or ChemBioDraw Ultra 13.0 (Perkin Elmer).
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. All combinations of the embodiments with respect to the chemical groups represented by the variables are specifically contemplated by the present invention and are disclosed herein to the extent that the combinations encompass compounds that produce stable compounds (i.e., compounds that can be isolated, characterized, and tested for biological activity) as if each and every combination were individually and explicitly disclosed. Moreover, all subcombinations of the chemical groups recited in the examples describing the variables are also specifically embraced by the present invention and are disclosed herein as if each and every subcombination of chemical groups were individually and specifically disclosed herein.
Chemical definition
The term "alkyl" refers to a straight or branched chain alkyl group having 1 to 12 carbon atoms in the chain. Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups herein that would be considered equivalent to any of the foregoing examples in light of the skilled artisan and the teachings provided herein.
The term "alkenyl" refers to a straight or branched chain hydrocarbon group having 2 to 12 carbon atoms in the chain and having one or more double bonds. Examples of alkenyl groups include vinyl, allyl, and but-3-en-1-yl. Included in this term are cis and trans isomers and mixtures thereof.
The term "alkynyl" refers to a straight or branched chain hydrocarbon group having 2 to 12 carbon atoms in the chain and having one or more triple bonds. Examples of alkynyl groups include ethynyl (-C.ident.CH) and propynyl (-CH) 2 C≡CH)。
The term "cycloalkyl" refers to a saturated or partially saturated, monocyclic or polycyclic carbocyclic ring having 3 to 12 ring atoms. Polycyclic carbocycles include fused, bridged and spiro polycyclic ring systems. Illustrative examples of cycloalkyl groups include the following entities in the form of suitable bonding moieties:
Figure GDA0003692253770000161
the term "halogen" represents chlorine, fluorine, bromine or iodine. The term "halo" represents chloro, fluoro, bromo or iodo.
The term "haloalkyl" refers to an alkyl group having one or more halo substituents, or one, two, or three halo substituents. Examples of haloalkyl groups include-CF 3 、-(CH 2 )F、-CHF 2 、-CH 2 Br、-CH 2 CF 3 and-CH 2 CH 2 F。
The term "aryl" refers to a group having 6 to 14 carbon atoms (C) with a completely conjugated pi-electron system 6 -C 14 ) All-carbon monocyclic or fused-ring polycyclic groups of (a). Aryl includes groups having 6 to 10 carbon atoms (e.g., "C 6-10 Aryl ") all-carbon monocyclic or fused-ring polycyclic. Examples of aryl groups are, but are not limited to, phenyl, naphthyl and anthracenyl. Aryl groups may be substituted or unsubstituted as described above for alkyl groups. Substituents also include those described elsewhere in this disclosure with respect to aryl groups.
The term "heterocycloalkyl" refers to a monocyclic or polycyclic structure that is saturated or partially saturated and has 3 to 12 ring atoms, wherein 1 to 5 ring atoms are selected from nitrogen, oxygen, and sulfur. Polycyclic ring systems include fused, bridged and spiro systems. The ring structure may optionally contain up to two oxo groups on carbon or sulfur ring members. Illustrative examples of heterocycloalkyl groups include the following entities in the form of suitable bonding moieties:
Figure GDA0003692253770000162
the term "heteroaryl" refers to a monocyclic, fused bicyclic, or fused polycyclic aromatic heterocycle having from 3 to 12 ring atoms per heterocycle (ring structures having ring atoms selected from carbon atoms and up to four heteroatoms selected from nitrogen, oxygen, and sulfur). Illustrative examples of heteroaryl groups include the following entities in the form of suitable bonding moieties:
Figure GDA0003692253770000171
"monocyclic" heteroaryl is an aromatic 5 or 6 membered heterocyclic ring. A 5-membered heteroaryl group contains up to four heteroatom ring atoms, wherein (a) one ring atom is oxygen and sulfur and 0,1 or 2 ring atoms are nitrogen, or (b) 0 ring atoms are oxygen or sulfur and up to 4 ring atoms are nitrogen. In some embodiments, the 5-membered heteroaryl is furan, thiophene, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, imidazole, oxadiazole, thiadiazole, triazole, or tetrazole. A6 membered heteroaryl group contains 1 or 2 nitrogen ring atoms. In some embodiments, the 6-membered heteroaryl is pyridine, pyrazine, pyrimidine, pyridazine, or triazine. A "bicyclic heteroaryl" is a fused bicyclic ring system comprising one heteroaryl ring fused to a phenyl or another heteroaryl ring.
The term "oxo" represents a carbonyl oxygen. For example, cyclopentyl substituted with oxo is cyclopentanone.
The term "substituted" is intended to mean that the specified group or moiety has one or more substituents. The term "unsubstituted" is intended to mean that the specified group has no substituents. When the term "substituted" is used to describe a structural system, the substitution is intended to occur at any position on the system where valence permits. In some embodiments, "substituted" is intended to mean that the specified group or moiety has 1,2, or 3 substituents. In other embodiments, "substituted" is intended to mean that the specified group or moiety has 1 or 2 substituents. In still other embodiments, "substituted" is intended to mean that the specified group or moiety has 1 substituent.
Any formula depicted herein is intended to represent the structural formula of the compound as well as certain variations or forms. For example, the formulae given herein are intended to include racemic forms, or one or more enantiomers, diastereomers, or geometric isomers, or mixtures thereof. In addition, any formula given herein is also intended to refer to a hydrate, solvate, or polymorph, or a mixture thereof, of such a compound.
Any formula given herein is also intended to represent unlabeled as well as isotopically labeled forms of the compounds. Isotopically labelled compoundsHaving a structure depicted by the formula (i) herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine and iodine, for example each 2 H、 3 H、 11 C、 13 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F、 36 Cl and 125 I. the isotopically-labeled compounds are useful in metabolic studies (preferably using 14 C) Reaction kinetics study (using, for example 2 H or 3 H) Detection or imaging techniques [ e.g. Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT)](including drug or substrate tissue distribution analysis) or radiotherapy of a patient. In addition, heavier isotopes are used (e.g. deuterium, i.e. 2 H) Substitution may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). Isotopically labeled compounds of the present invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
Nomenclature "(ATOM) i-j And j>i as applied to a class of substituents herein is intended to refer to the following examples of the invention: each and every one of the carbon member numbers from i to j (inclusive) is independently implemented. For example, the term C 1-3 Independently means having one carbon member (C) 1 ) Example of (5), having two carbon members (C) 2 ) Examples of (D) and (D) having three carbon members (C) 3 ) Examples of (1).
Any disubstituted group mentioned herein is intended to encompass a wide range of such attachment possibilities when more than one is allowed. For example, reference to a disubstituent-a-B- (wherein a ≠ B) refers herein to the disubstituent with a attached to a first substituted member and B attached to a second substituted member, and it also refers to the disubstituent with a attached to a second member and B attached to the first substituted member.
The invention also includes pharmaceutically acceptable salts of the compounds represented by formula (I) or (I-a), preferably those described above and the specific compounds exemplified herein, and pharmaceutical compositions comprising and methods of using such salts.
"pharmaceutically acceptable salt" is intended to mean a salt of a free acid or base of a compound represented herein that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to a subject. See generally, s.m. bellhe (s.m. berge) et al, "Pharmaceutical Salts (Pharmaceutical Salts)," journal of medical science (j.pharm.sci., 1977,66,1-19. Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of an individual without undue toxicity, irritation, or allergic response. The compounds described herein can have sufficiently acidic groups, sufficiently basic groups, two types of functional groups, or more than one of each type, and thus react with many inorganic or organic bases and inorganic and organic acids to form pharmaceutically acceptable salts.
Examples of pharmaceutically acceptable salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, hydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, borate, nitrate, propionate, caprate, caprylate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1, 4-dioate, hexyne-1, 6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, sulfonate, methanesulfonate, propanesulfonate, toluenesulfonate, xylenesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, γ -hydroxybutyrate, glycolate, tartrate and mandelate. A list of other suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 17 th edition, merck Publishing Company (Mack Publishing Company), iston, pa., 1985.
For compounds of formula (I) or (I-a) containing a basic nitrogen, pharmaceutically acceptable salts may be prepared by any suitable method available in the art, for example by treating the free base with: inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like; or organic acids such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, pyranosyl acids (pyranosidyl acids) (e.g., glucuronic or galacturonic acids), alpha-hydroxy acids (e.g., mandelic, citric or tartaric acids), amino acids (e.g., aspartic or glutamic acid), aromatic acids (e.g., benzoic, 2-acetoxybenzoic, naphthoic or cinnamic acids), sulfonic acids (e.g., laurylsulfonic, p-toluenesulfonic, methanesulfonic or ethanesulfonic acid), or any compatible mixtures of acids such as those given by way of example herein, and any other acid and mixtures thereof deemed equivalent or acceptable alternatives according to the ordinary level of skill in the art.
The invention also relates to pharmaceutically acceptable prodrugs of compounds of formula (I) or (I-A) and methods of treatment employing the pharmaceutically acceptable prodrugs. The term "prodrug" is intended to mean a precursor of a specified compound that, upon administration to a subject, produces the compound in vivo via a chemical or physiological process (e.g., solvolysis or enzymatic cleavage) or under physiological conditions (e.g., a prodrug at physiological pH is converted to a compound of formula (I) or (I-a)). A "pharmaceutically acceptable prodrug" is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject. An illustrative procedure for selecting and preparing suitable prodrug derivatives is described, for example, in "prodrug Design (Design of produgs)", editions h. Bindgard (h. Bundgaard), eisavir (Elsevier), 1985.
The invention also relates to pharmaceutically active metabolites of compounds of formula (I) or (I-A) and the use of said metabolites in the methods of the invention. "pharmaceutically active metabolite" is intended to mean the pharmacologically active product of the metabolism in vivo of a compound of formula (I) or (I-A) or a salt thereof. Prodrugs and active metabolites of a compound may be determined using conventional techniques known or available in the art. See, e.g., betrolini (bertoni), et al, journal of medicinal chemistry 1997,40,2011-2016; shan (Shan) et al, J.Med.Sci 1997,86 (7), 765-767; the Baggen zodiac, drug development research (Drug Dev. Res.) 1995,34,220-230; boudor (Bodor), progress in drug research (adv. Drug res.) 1984,13,255-331; poddard, design of Prodrugs (Design of produgs) (Esrivier Press, 1985); and Larsen (Larsen), design and Application of Prodrugs, drug Design and Development (Design and Application of produgs, drug Design and Development) (edited by Krogsgaard-Larsen et al, harwood Academic Publishers, 1991).
Representative examples
In some embodiments of formula (I-a), ring a 'is monocyclic aryl or heteroaryl and ring B' is bicyclic heteroaryl. In other embodiments, ring a 'is a bicyclic heteroaryl and ring B' is a monocyclic aryl or heteroaryl. In some embodiments, ring a' is phenyl or 6 membered heteroaryl. In other embodiments, ring B' is a bicyclic heteroaryl containing 1,2, or 3 nitrogen ring atoms. In other embodiments, ring a' is phenyl or pyridyl.
In yet other embodiments, ring a' is phenyl. In still other embodiments, - (R) 3’ ) p’ Substituted ring A' is
Figure GDA0003692253770000201
In still other embodiments, - (R) 3’ ) p’ Substituted ring A' is
Figure GDA0003692253770000202
In some embodiments, ring B' is:
Figure GDA0003692253770000203
wherein Z 1 -Z 7 As defined herein. In still other embodiments, ring B' is:
Figure GDA0003692253770000204
wherein Z 1-7 As defined elsewhere herein. In still other embodiments, ring B' is:
Figure GDA0003692253770000205
in still other embodiments, ring B' is
Figure GDA0003692253770000206
In still other embodiments, ring B' is
Figure GDA0003692253770000207
In other embodiments of formula (I-a), ring a' is a bicyclic heteroaryl and is:
Figure GDA0003692253770000211
wherein Z 1 -Z 7 As defined herein. In still other embodiments, ring a' is:
Figure GDA0003692253770000212
wherein Z 1-7 As defined elsewhere herein. In still other embodiments, ring a' is:
Figure GDA0003692253770000213
in still other embodiments, ring A' is
Figure GDA0003692253770000214
In still other embodiments, ring A' is
Figure GDA0003692253770000215
In some embodiments, ring B' is a monocyclic aryl or heteroaryl. In other embodiments, ring B' is phenyl. In other embodiments, ring a' is pyridyl.
In some embodiments, each R is 3’ Independently deuterium, fluoro, chloro, bromo, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN, -CF 3 、-NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 C 1-4 Alkyl, -CO 2 H、-NHC(O)C 1-4 Alkyl, -SO 2 C 1-4 Alkyl, -C (O) NH 2 、-C(O)NH(C 1-4 Alkyl), -C (O) N (C) 1-4 Alkyl radical) 2 Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholinyl. In still other embodiments, each R 3’ Independently fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3 . In still other embodiments, each R 3’ Is fluorine or chlorine.
In some embodiments, R 7’ Is H, deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furyl, thiofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl or monocyclic heteroaryl, each of which is substituted or unsubstituted as in formula (I-A). In other embodiments, R 7’ Is H, or is methyl, ethyl, propyl, isopropyl or cyclopropyl, each unsubstituted or substituted as in formula (I-A). In yet other embodiments, R7' is H or methyl or ethyl, each of which is unsubstituted or substituted as follows: halogen, -OH, -OC 1-4 Alkyl and substituted benzeneNH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 Cycloalkyl or monocyclic heterocycloalkyl. In still other embodiments, R 7’ Is H, methyl, hydroxyethyl, -CH 2 CONH 2 Or 3-pyrrolidinylmethyl. In still other embodiments, R 7’ Is H or methyl.
In some embodiments, R 1’ And R 2’ Each independently is H, deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl, or monocyclic heteroaryl, each of which is substituted or unsubstituted as in formula (I-A). In other embodiments, R 1’ Is H. In still other embodiments, R 2’ Deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl or monocyclic heteroaryl, each of which is substituted or unsubstituted as in formula (I-a). In still other embodiments, R 2’ Is H or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 Cycloalkyl or monocyclic heterocycloalkyl. In still other embodiments, R 2’ Is H, methyl, fluoromethyl, hydroxymethyl or cyclopropyl. In still other embodiments, R 2’ Is H. In still other embodiments, R 2’ Is a methyl group.
In some embodiments, each R is k’ Independently H, methyl, ethyl, propyl, isopropyl or cyclopropyl. In other embodiments, each R k’ Independently is H or methyl.
In some embodiments, each L 1 And L 2 Independently is-CH 2 -or-CH (methyl) -, -CH (substituted methyl) -, -CH (C) 3-6 Cyclopropyl) -, -CH (OH))-、-O-、-NH-、-N(C 1-4 Alkyl) -, -N (C) 3-6 Cyclopropyl) -, -S (O) -or-SO 2 -. In some embodiments, - (L) 1 ) n’ is-CH 2 -O-、-CH(C 1-4 Alkyl) -O-or-CH (C) 3-6 Cycloalkyl) -O-. In other embodiments, - (L) 1 ) n’ -is-CH (H or optionally substituted C) 1-4 Alkyl) -N (H or optionally substituted C 1-4 Alkyl) -, -CH (CO) 2 C 1-4 Alkyl or C (O) N (H or C) 1-4 Alkyl radical) 2 ) -N (H or optionally substituted C) 1-4 Alkyl groups). In still other embodiments, - (L) 1 ) n’ is-CH 2 S(O) 0-2 -. In other embodiments, - (L) 1 ) n’ is-SO 2 -N (H or C) 1-4 Alkyl). In some embodiments, - (L) 1 ) n’ Is- (CH) 2 ) 3 -. In some embodiments, - (L) 1 ) n’ Is- (CH) 2 ) 2 -. In some embodiments, - (L) 1 ) n’ -is-CH (CH) 3 )CH 2 -。
In some embodiments, - (L) 2 ) m’ is-O- (C (R) 1’ )(R 2’ )) 2-3 -. In other embodiments, - (L) 2 ) m’ is-O- (CH) 2 ) 2-3 -. In other embodiments, - (L) 2 ) m’ is-N (R) k’ )-(C(R 1’ )(R 2’ )) 2-3 -. In other embodiments, - (L) 2 ) m’ is-N (H or C) 1-4 Alkyl group) - (CH 2 ) 2-3 -. In other embodiments, - (L) 2 ) m’ is-S- (C (R) 1’ )(R 2’ )) 2-3 -. In other embodiments, - (L) 2 ) m’ is-SO 2 -(C(R 1’ )(R 2’ )) 2-3 -. In still other embodiments, - (L) 2 ) m’ is-SO 2 -N(R k’ )-(C(R 1’ )(R 2’ )) 2 -. In still other embodiments, - (L) 2 ) m’ Is- (C (R) 1’ )(R 2’ )) 3 -。
In some embodiments, m' is 3. In other embodiments, m' is 4. In still other embodiments, m' is 5. In some embodiments, n' is 2. In other embodiments, n' is 3. In still other embodiments, n' is 4. In some embodiments, p' is 0,1, or 2. In other embodiments, p' is 1 or 2. In some embodiments, q' is 0. In other embodiments, q' is 1. In still other embodiments, q' is 2.
In some embodiments, formula (I-A) is a compound of formula (I) or a pharmaceutically acceptable salt thereof. In other embodiments, the compound of formula (I-a) is a compound of formula (I), wherein each variable is independently defined as shown below for formula (I). In some embodiments, the variables of formula (I-a) are mapped on formula (I) as follows: a' is A; b' is B; r 1’ Is R 1 ;R 2’ Is R 2 ;R 3’ Is R 3 ;R 4’ Is R 4 ;R 7’ Is R 7 ;R a’ -R f’ And R i’ -R k’ Respectively mapped to R a -R f And R i -R k The above step (1); and L is 1 And L 2 Are respectively-Y- (C (R) 5 )(R 6 )) m -and-C ((R) 1 )(R 2 )) n -X-。
In some embodiments of formula (I), ring a is phenyl or 6 membered heteroaryl. In other embodiments, ring a is phenyl or pyridyl. In still other embodiments, ring a is phenyl. In still other embodiments, - (R) 3 ) p Substituted ring A is
Figure GDA0003692253770000231
In still other embodiments, - (R) 3 ) p Substituted ring A is
Figure GDA0003692253770000232
In some embodiments, each R is 3 Independently deuterium, fluoro, chloro, bromo, methyl, ethyl, propyl, isopropyl, methoxy, ethoxyRadical, isopropoxy, -CN, -CF 3 、-NH 2 、-NH(C 1-4 Alkyl) -, -N (C) 1-4 Alkyl radical) 2 、-CO 2 C 1-4 Alkyl, -CO 2 H、-NHC(O)C 1-4 Alkyl, -SO 2 C 1-4 Alkyl, -C (O) NH 2 、-C(O)NH(C 1-4 Alkyl), -C (O) N (C) 1-4 Alkyl radical) 2 Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholinyl. In still other embodiments, each R 3 Independently is fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3 . In still other embodiments, each R 3 Is fluorine or chlorine.
In still other embodiments, - (R) 3 ) p Substituted ring A is
Figure GDA0003692253770000233
Wherein R is 3a And R 3b Each independently is H, fluoro or chloro, and M is CH or N. In some embodiments, R 3a Is fluorine.
In some embodiments, p is 1 or 2. In other embodiments, p is 0. In still other embodiments, p is 1. In still other embodiments, p is 2.
In some embodiments, ring B is a bicyclic heteroaryl. In other embodiments, ring B is a 9-membered bicyclic heteroaryl.
In some embodiments, each R is 4 Independently deuterium, fluoro, chloro, bromo, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN, -CF 3 、-NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 C 1-4 Alkyl, -CO 2 H、-NHC(O)C 1-4 Alkyl, -SO 2 C 1-4 Alkyl, -C (O) NH 2 、-C(O)NH(C 1-4 Alkyl), -C (O) N (C) 1-4 Alkyl radical) 2 Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl or thiomorpholinyl. In still other embodiments, each R 4 Independently is fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3
In other embodiments, - (R) 4 ) q Substituted ring B is:
Figure GDA0003692253770000241
wherein Z 1 、Z 2 、Z 3 And Z 6 Each independently is-C (R) x ) -or N;
wherein each R is x Independently of each other is H, deuterium, halogen, C 1-4 Alkyl, -O-C 1-4 Alkyl, -OH, -NH 2 、-NHC 1-4 Alkyl, -NH-phenyl, -NH-heteroaryl, CN or-CF 3
Z 4 And Z 5 Each independently is-C-or-N-; and is
Z 7 is-CH-, -N-or-NH-.
In other embodiments:
(a)Z 1 、Z 4 and Z 7 Each is-N-;
(b)Z 1 、Z 5 and Z 7 Each is-N-;
(c)Z 1 and Z 3 Each is-N-and Z 7 is-NH-;
(d)Z 3 is-N-and Z 7 is-NH-;
(e)Z 3 and Z 6 Each is-N-and Z 7 is-NH-;
(f)Z 2 、Z 4 and Z 7 Each is-N-;
(g)Z 1 、Z 2 、Z 4 and Z 7 Each is-N-;
(h)Z 1 、Z 3 and Z 4 Each is-N-;
(i)Z 3 and Z 4 Each is-N-;
(j)Z 1 、Z 2 、Z 5 and Z 7 Each is-N-;
(k)Z 2 、Z 5 and Z 7 Each is-N-;
(l)Z 3 and Z 5 Each is-N-;
(m)Z 3 、Z 5 and Z 6 Each is-N-;
(n)Z 1 、Z 5 、Z 6 and Z 7 Each is-N-;
(o)Z 2 、Z 5 、Z 6 and Z 7 Each is-N-; or
(p)Z 1 、Z 3 And Z 6 Each is-N-and Z 7 is-NH-.
In still other embodiments of (a) - (p), each Z ring atom not explicitly defined is independently-C-or-C (R) x ) - (consistent with the definition of the ring atoms). In still other embodiments, Z 3 is-N-. In other embodiments, Z 7 is-N-or-NH-. In still other embodiments, Z 3 is-N-and Z 7 is-N-or-NH-. In still other embodiments, - (R) 4 ) q Substituted ring B is:
Figure GDA0003692253770000251
wherein Z 1-7 Are otherwise defined as described above.
In still other embodiments, - (R) 4 ) q Substituted ring B is:
Figure GDA0003692253770000252
in still other embodiments, - (R) 4 ) q Substituted ring B is
Figure GDA0003692253770000253
In still other embodiments, - (R) 4 ) q Substituted ring B is
Figure GDA0003692253770000254
In some embodiments, q is 0. In other embodiments, q is 1.
In some embodiments, R 1 And R 2 Each independently is H, deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl, or monocyclic heteroaryl, each as substituted or unsubstituted in formula (I). In other embodiments, R 1 Is H. In still other embodiments, R 2 Deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl or monocyclic heteroaryl, each as substituted or unsubstituted in formula (I). In still other embodiments, R 2 Is H or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 Cycloalkyl or monocyclic heterocycloalkyl. In still other embodiments, R 2 Is H, methyl, fluoromethyl, hydroxymethyl or cyclopropyl. In still other embodiments, R 2 Is H. In still other embodiments, R 2 Is methyl. In still other embodiments, R 1 Is H, and R 2 Is not H and has the stereochemical configuration shown below:
Figure GDA0003692253770000261
in still other embodiments, R 1 And R 2 Together form C 3-6 A cycloalkyl group. In other embodiments, R 1 And R 2 Together form an optional ground warp C 1-4 Alkyl-substituted 5 or 6 membered heterocycloalkyl.
In some embodiments, n is 1 or 2. In still other embodiments, n is 1.
In some embodiments, R 5 And R 6 Each independently is H, deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl, or monocyclic heteroaryl, each as substituted or unsubstituted in formula (I). In other embodiments, each R 5 Is H. In still other embodiments, each R 6 Independently H, or methyl, ethyl or cyclopropyl, each as substituted or unsubstituted in formula (I). In still other embodiments, each R 6 Independently H or unsubstituted or-OH substituted methyl. In still other embodiments, each R 6 Is H or methyl. In still other embodiments, R 5 And R 6 Together form C 3-6 A cycloalkyl group. In other embodiments, R 5 And R 6 Together form an optional ground warp C 1-4 Alkyl-substituted 5 or 6 membered heterocycloalkyl.
In some embodiments, m is 2 or 3. In other embodiments, m is 2.
In some embodiments, R 7 Is H, deuterium, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, pyrrolidinyl, furanyl, thiofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl or monocyclic heteroaryl, each as substituted or unsubstituted in formula (I). In other embodiments, R 7 Is H, or is methyl, ethyl, propyl, isopropyl or cyclopropyl, each of which is unsubstituted or substituted as in formula (I). In still other embodiments, R 7 Is H or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 Cycloalkyl or monocyclic heterocycloalkyl. In still other embodiments, R 7 Is H, methyl, hydroxyethyl, -CH 2 CONH 2 Or 3-pyrrolidinylmethyl. In still other embodiments, R 7 Is H or methyl.
In some embodiments, each of X and Y is independently-O-or-N (R) k ) -. In some embodiments, X is-O-or-N (R) k ) -. In some embodiments, Y is-O-. In some embodiments, each R is k Independently H, methyl, ethyl, propyl, isopropyl or cyclopropyl. In other embodiments, each R k Independently is H or methyl.
In some embodiments, the compound of formula (I) or (I-a) is a compound of formula (II):
Figure GDA0003692253770000271
wherein M and R 3 、q、R 2 、X、R 7 And Z 1-7 Each defined in any of the ways listed above;
R 5a 、R 5b 、R 6a and R 6b Each is R 5 And R is 6 As defined above in any one of the several ways listed;
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of formula (I) or (I-a) is a compound of formula (III):
Figure GDA0003692253770000272
wherein
M is CH or N;
R 3a and R 3b Each independently of the other being H, fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3
R 2a Is H or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 Cycloalkyl or monocyclic heterocycloalkyl;
X 1 is O or-N (CH) 3 )-;
R 5a 、R 6a 、R 5b And R 6b Each independently is H, or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 Cycloalkyl or monocyclic heterocycloalkyl;
R 7a is H or methyl or ethyl, each of which is unsubstituted or substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 Cycloalkyl or monocyclic heterocycloalkyl;
Z 1-7 each defined in any of the ways listed above;
or a pharmaceutically acceptable salt thereof.
In some embodiments of formula (III), M is CH.
In other embodiments, R 3a And R 3b Each independently is H, fluoro or chloro. In still other embodiments, R 3a Is H or fluorine. In still other embodiments, R 3a Is fluorine. In still other embodiments, R 3b Is H or chlorine.
In some embodiments of formula (III), R 2a Is H, methyl, fluoromethyl or cyclopropyl.
In some embodiments of formula (III), X 1 Is O. In other embodiments, X is-N (CH) 3 )-。
In some embodiments, R 7a Is H, methyl, hydroxyethyl,-CH 2 CONH 2 Or 3-pyrrolidinylmethyl. In other embodiments, R 7a Is H or methyl.
In some embodiments, the compound of formula (I) or (I-a) is a compound of formula (IV):
Figure GDA0003692253770000281
wherein
M is CH or N;
X 1 and X 1 ' independently is-C (R) 1a )(R 2a )-、-S-、-S(O)-、-S(O) 2 -, -O-or-N (R) k’ )-;
Each R 1a And R 2a Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 6-10 Aryl, -C (O) OR a’ 、-C(O)NR a’ R b’ 、-NR a’ R b’ 、-SR a’ 、-S(O)R a’ 、-S(O)NR a’ 、-S(O) 2 R a’ 、-S(O) 2 NR a’ OR-OR a’ In which C is 1-6 Each hydrogen atom in the alkyl group is independently optionally substituted by: deuterium, halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、NHC(O)C 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) C 1-4 Alkyl, -NHC (O) NHC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) NHC 1-4 Alkyl, NHC (O) N (C) 1-4 Alkyl radical) 2 、-N(C 1-4 Alkyl) C (O) N (C) 1-4 Alkyl radical) 2 、-NHC(O)OC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) OC 1-4 Alkyl, -CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 、-SC 1-4 Alkyl, -S (O) C 1-4 Alkyl, -S (O) 2 C 1-4 Alkyl, -S (O) NH (C) 1-4 Alkyl), -S (O) 2 NH(C 1-4 Alkyl), -S (O) N (C) 1-4 Alkyl radical) 2 、-S(O) 2 N(C 1-4 Alkyl radical) 2 、C 3-6 Cycloalkyl or 3 to 7 membered heterocycloalkyl;
R 3a and R 3b Each independently H, deuterium, fluoro, chloro, bromo, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3
R 7a Is H, C 1-6 Alkyl or 3 to 7 membered heterocycloalkyl, wherein C 1-6 Each hydrogen atom in alkyl or 3-to 7-membered heterocycloalkyl is independently optionally substituted by: deuterium, halogen, -CN, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 Cycloalkyl or monocyclic heterocycloalkyl;
each R k’ Independently of each other H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl; wherein R is k ' C in 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in an aryl or a monocyclic or bicyclic heteroaryl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl OR-OR a’
Wherein each R is a’ And R b’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or heteroaryl;
each Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 Independently N, NH or C (R) x ) Wherein each R is x When present, is independently H, deuterium, halogen, C 1-4 Alkyl, -O-C 1-4 Alkyl, -OH, -NH 2 、-NH(C 1-4 Alkyl), -NH (phenyl), -NH (heteroaryl), CN or-CF 3 Provided that Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 At least one of which is N or NH; and is
m' is 2 or 3;
or a pharmaceutically acceptable salt thereof.
In some embodiments, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 And Z 3 Is N, Z 7 Is NH and Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 6 Is N, Z 7 Is NH and Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 Is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 6 Is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 4 And Z 7 Is N and Z 1 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 And Z 7 Is N and Z 2 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 4 And Z 7 Is N and Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 5 And Z 7 Is N and Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 、Z 5 And Z 6 Is N and Z 1 、Z 2 、Z 4 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 、Z 6 And Z 7 Is N and Z 2 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 And Z 4 Is N and Z 3 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 4 Is N and Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 4 Is N and Z 1 、Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 And Z 7 Is N and Z 1 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 5 Is N and Z 1 、Z 2 、Z 4 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 、Z 6 And Z 7 Is N and Z 1 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H.
In some embodiments, R k’ Selected from the group consisting of: H. methyl, ethyl, propyl, isopropyl, cyclopropyl, 2-hydroxyethyl, 2-hydroxy-2-methyl-propylAnd N-methyl-pyrrol-3-yl. In some embodiments, M is CH. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, and X 1 is-N (R) k’ ) -. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-N (R) k’ ) -, and X 1 ' is-O-. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-C (R) 1a )(R 2a ) -, and X 1 ' is-O-.
In some embodiments, the compound of formula (I) or (I-a) is a compound of formula (V):
Figure GDA0003692253770000301
wherein
M is CH or N;
X 1 and X 1 ' independently is-C (R) 1a )(R 2a )-、-S-、-S(O)-、-S(O) 2 -, -O-or-N (R) k’ )-;
Each R 1a And R 2a Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 6-10 Aryl, -C (O) OR a’ 、-C(O)NR a’ R b’ 、-NR a’ R b’ 、-SR a’ 、-S(O)R a’ 、-S(O)NR a’ 、-S(O) 2 R a’ 、-S(O) 2 NR a’ OR-OR a’ In which C is 1-6 Each hydrogen atom in the alkyl group is independently optionally substituted by: deuterium, halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、NHC(O)C 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) C 1-4 Alkyl, -NHC (O) NHC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) NHC 1-4 Alkyl, NHC (O) N (C) 1-4 Alkyl radical) 2 、-N(C 1-4 Alkyl) C (O) N (C) 1-4 Alkyl radical) 2 、-NHC(O)OC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) OC 1-4 Alkyl, -CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 、-SC 1-4 Alkyl, -S (O) C 1-4 Alkyl, -S (O) 2 C 1-4 Alkyl, -S (O) NH (C) 1-4 Alkyl), -S (O) 2 NH(C 1-4 Alkyl), -S (O) N (C) 1-4 Alkyl radical) 2 、-S(O) 2 N(C 1-4 Alkyl radical) 2 、C 3-6 Cycloalkyl or 3 to 7 membered heterocycloalkyl;
R 3a and R 3b Each independently of the other being H, fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3
R 7a Is H, C 1-6 Alkyl or 3 to 7 membered heterocycloalkyl wherein C 1-6 Each hydrogen atom in alkyl or 3-to 7-membered heterocycloalkyl is independently optionally substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 Cycloalkyl or monocyclic heterocycloalkyl;
each R k’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 A cycloalkyl group, a,3 to 7 membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl; wherein R is k ' C in 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in an aryl or a monocyclic or bicyclic heteroaryl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl OR-OR a’
Wherein each R is a’ And R b’ Independently of each other is H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or heteroaryl;
each Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 Independently N, NH or C (R) x ) Wherein each R is x When present, is independently H, deuterium, halogen, C 1-4 Alkyl, -O-C 1-4 Alkyl, -OH, -NH 2 、-NH(C 1-4 Alkyl), -NH (phenyl), -NH (heteroaryl), CN or-CF 3 Provided that Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 At least one of which is N or NH; and is
m' is 2 or 3;
or a pharmaceutically acceptable salt thereof.
In some embodiments, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 And Z 3 Is N, Z 7 Is NH and Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 6 Is N, Z 7 Is NH and Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments of the present invention, the,Z 3 is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 6 Is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 4 And Z 7 Is N and Z 1 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 And Z 7 Is N and Z 2 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 4 And Z 7 Is N and Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 5 And Z 7 Is N and Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 、Z 5 And Z 6 Is N and Z 1 、Z 2 、Z 4 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 、Z 6 And Z 7 Is N and Z 2 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 And Z 4 Is N and Z 3 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 4 Is N and Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 4 Is N and Z 1 、Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 And Z 7 Is N and Z 1 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 5 Is N and Z 1 、Z 2 、Z 4 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 、Z 6 And Z 7 Is N and Z 1 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H.
In some embodiments, R k’ Selected from the group consisting of: H. methyl, ethyl, propyl, isopropyl, cyclopropyl, 2-hydroxyethyl, 2-hydroxy-2-methyl-propyl and N-methyl-pyrrol-3-yl. In some embodiments, M is CH. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, and X 1 is-N (R) k’ ) -. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-N (R) k’ ) -, and X 1 ' is-O-. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-C (R) 1a )(R 2a ) -, and X 1 ' is-O-.
In some embodiments, the compound of formula (I) or (I-a) is a compound selected from the group consisting of:
Figure GDA0003692253770000321
Figure GDA0003692253770000331
wherein
M is CH or N;
X 1 and X 1 ' independently is-C (R) 1a )(R 2a )-、-S-、-S(O)-、-S(O) 2 -, -O-or-N (R) k’ )-;
Each R 1a And R 2a Independently of each other H, deuterium, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 6-10 Aryl, -C (O) OR a’ 、-C(O)NR a’ R b’ 、-NR a’ R b’ 、-SR a’ 、-S(O)R a’ 、-S(O)NR a’ 、-S(O) 2 R a’ 、-S(O) 2 NR a’ OR-OR a’ In which C is 1-6 Each hydrogen atom in the alkyl group is independently optionally substituted by: deuterium, halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、NHC(O)C 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) C 1-4 Alkyl, -NHC (O) NHC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) NHC 1-4 Alkyl, NHC (O) N (C) 1-4 Alkyl radical) 2 、-N(C 1-4 Alkyl) C (O) N (C) 1-4 Alkyl radical) 2 、-NHC(O)OC 1-4 Alkyl, -N (C) 1-4 Alkyl) C (O) OC 1-4 Alkyl, -CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 、-SC 1-4 Alkyl, -S (O) C 1-4 Alkyl, -S (O) 2 C 1-4 Alkyl, -S (O) NH (C) 1-4 Alkyl), -S (O) 2 NH(C 1-4 Alkyl), -S (O) N (C) 1-4 Alkyl radical) 2 、-S(O) 2 N(C 1-4 Alkyl radical) 2 、C 3-6 Cycloalkyl or 3 to 7 membered heterocycloalkyl;
R 3a and R 3b Each independently of the others being H, fluorine, chlorine, bromine, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, -CN or-CF 3
R 7a Is H, C 1-6 Alkyl or 3 to 7 membered heterocycloalkyl, wherein C 1-6 Each hydrogen atom in alkyl or 3-to 7-membered heterocycloalkyl is independently optionally substituted by: halogen, -OH, -OC 1-4 Alkyl, -NH 2 、-NH(C 1-4 Alkyl), -N (C) 1-4 Alkyl radical) 2 、-CO 2 H、-CO 2 C 1-4 Alkyl, -CONH 2 、-CONH(C 1-4 Alkyl), -CON (C) 1-4 Alkyl radical) 2 Cycloalkyl or monocyclic heterocycloalkyl;
each R k’ Independently of each other H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or monocyclic or bicyclic heteroaryl; wherein R is k ' C in 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Each hydrogen atom in the aryl or monocyclic or bicyclic heteroaryl is independently optionally substituted by: deuterium, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl OR-OR a’
Wherein each R is a’ And R b’ Independently of each other H, deuterium, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-6 Cycloalkyl, 3-to 7-membered heterocycloalkyl, C 6-10 Aryl or heteroaryl;
each Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 Independently N, NH or C (R) x ) Wherein each R is x When present, is independently H, deuterium, halogen, C 1-4 Alkyl, -O-C 1-4 Alkyl, -OH, -NH 2 、-NH(C 1-4 Alkyl), -NH (phenyl), -NH (heteroaryl), CN or-CF 3 Provided that Z is 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 Or Z 7 At least one of which is N or NH; and is
m' is 2 or 3;
or a pharmaceutically acceptable salt thereof.
In some embodiments, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 And Z 3 Is N, Z 7 Is NH and Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 6 Is N, Z 7 Is NH and Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 Is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 6 Is N, Z 7 Is NH and Z 1 、Z 2 、Z 4 And Z 5 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 4 And Z 7 Is N and Z 1 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 And Z 7 Is N and Z 2 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 4 And Z 7 Is N and Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 、Z 5 And Z 7 Is N and Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 、Z 5 And Z 6 Is N and Z 1 、Z 2 、Z 4 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 5 、Z 6 And Z 7 Is N and Z 2 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 2 And Z 4 Is N and Z 3 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 1 、Z 3 And Z 4 Is N and Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 4 Is N and Z 1 、Z 2 、Z 5 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 And Z 7 Is N and Z 1 、Z 3 、Z 4 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 3 And Z 5 Is N and Z 1 、Z 2 、Z 4 、Z 6 And Z 7 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, Z 2 、Z 5 、Z 6 And Z 7 Is N and Z 1 、Z 3 And Z 4 Is C (R) x ) Wherein each R is x When present is H.
In some embodiments, R k’ Selected from the group consisting of: H. methyl, ethyl, propyl, isopropyl, cyclopropyl, 2-hydroxyethyl, 2-hydroxy-2-methyl-propyl and N-methyl-pyrrol-3-yl. In some embodiments, M is CH. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N and Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, and X 1 is-N (R) k’ ) -. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-N (R) k’ ) -, and X 1 ' is-O-. In some embodiments, M is CH, Z 1 、Z 4 And Z 7 Is N, Z 2 、Z 3 、Z 5 And Z 6 Is C (R) x ) Wherein each R is x When present is H, X 1 is-C (R) 1a )(R 2a ) -, and X 1 ' is-O-.
In other embodiments, the compound of formula (I) or (I-a) is selected from the group consisting of: (13R) -5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; 5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-modified-4 (5H) -one; (13R) -11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (13R) -12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenylpyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; 12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenylpolypyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -12-chloro-11-fluoro-5- (2-hydroxyethyl) -13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 12-chloro-11-fluoro-5- (2-hydroxyethyl) -13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; 2- [ (13R) -12-chloro-11-fluoro-13-methyl-4-oxo-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-ring-5 (4H) -yl ] acetamide; 2- [ 12-chloro-11-fluoro-13-methyl-4-oxo-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-5 (4H) -yl ] acetamide; (13R) -12-chloro-11-fluoro-13-methyl-5- (pyrrolidin-2-ylmethyl) -6, 7-dihydro-13H-1, 15-ethenylpyrrolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 12-chloro-11-fluoro-13-methyl-5- (pyrrolidin-2-ylmethyl) -6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -12-chloro-11-fluoro-7- (hydroxymethyl) -5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 12-chloro-11-fluoro-7- (hydroxymethyl) -5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (13S) -11-fluoro-13- (fluoromethyl) -5-methyl-6, 7-dihydro-13H-1, 15-ethenylpyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; 11-fluoro-13- (fluoromethyl) -5-methyl-6, 7-dihydro-13H-1, 15-ethenylpyrrolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; (13R) -13-cyclopropyl-11-fluoro-5-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 13-cyclopropyl-11-fluoro-5-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (13R) -11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; 11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-form-4 (5H) -one; (13R) -12-chloro-11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 12-chloro-11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; 12-chloro-11-fluoro-6-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; 12-chloro-11-fluoro-7-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (8R) -9-chloro-10-fluoro-8-methyl-15, 16-dihydro-8H-3, 6-ethenylimidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; 9-chloro-10-fluoro-8-methyl-15, 16-dihydro-8H-3, 6-vinylimidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; (7R) -8-chloro-9-fluoro-7-methyl-14, 15-dihydro-2h, 7H-3,5- (nitrilomethylene bridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine cyclol-16 (13H) -one; 8-chloro-9-fluoro-7-methyl-14, 15-dihydro-2h, 7h-3,5- (nitrilomethylene bridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine slow-ring-16 (13H) -one; (5R) -3-fluoro-5-methyl-14, 15-dihydro-5H, 10H-9,7- (nitrilomethylene bridging) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,10,3,7] dioxadiazidenyl ring-12 (13H) -one; 3-fluoro-5-methyl-14, 15-dihydro-5H, 10H-9,7- (nitrilomethylenebridge) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,10,3,7] dioxadiazidetridec-12 (13H) -one; (5R) -3-fluoro-5, 16-dimethyl-13, 14,15, 16-tetrahydro-5H-9, 7- (nitrilomethylene bridging) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,3,7,10] oxatrinexazacyclol-12 (10H) -one; 3-fluoro-5, 16-dimethyl-13, 14,15, 16-tetrahydro-5H-9, 7- (nitrilomethylenebridge-yl) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,3,7,10] oxa-triazatridean-12 (10H) -one; (13R) -12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-2H, 13H-1,15- (nitrilomethylenebridge) pyrrolo [3,4-f ] [1,10,4] benzodioxazepin-4 (5H) -one; 12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-2h, 13h-1,15- (nitrilomethylenebridge) pyrrolo [3,4-f ] [1,10,4] benzodiazepine-4 (5H) -one; (7R) -8-chloro-9-fluoro-7, 15-dimethyl-14, 15-dihydro-2h, 7H-3,5- (nitrilomethylenebridge) pyrazolo [3,4-f ] [1,10,4] benzodiazepine-16 (13H) -one; 8-chloro-9-fluoro-7, 15-dimethyl-14, 15-dihydro-2h, 7h-3,5- (nitrilomethylene bridging) pyrazolo [3,4-f ] [1,10,4] benzodiazepine-ring-16 (13H) -one; 11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (13R) -12-chloro-11-fluoro-13, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecane-4 (5H) -one; 12-chloro-11-fluoro-13, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 12-chloro-11-fluoro-5, 14-dimethyl-6, 7,13, 14-tetrahydro-15, 1- (nitrilomethylenebridging) pyrazolo [4,3-f ] [1,4,10] benzoxadiazidenyl ring-4 (5H) -one; 12-chloro-11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-15, 1- (nitrilomethylenebridging) pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 12-chloro-11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15- (nitrilomethylenebridge) pyrrolo [3,2-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 12-chloro-11-fluoro-14-methyl-6,7,13,14-tetrahydro-1, 15- (nitrilomethylene-bridge) pyrrolo [3,2-f ] [1,4,10] benzoxadiazepin-4 (5H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-3, 6-ethenylimidazo [5,1-f ] [1,4,7,8,10] benzoxatetrazetidin-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-6, 3- (nitrilomethylenebridge) imidazo [5,1-f ] [1,4,7,8,10] benzoxatetraazatridec-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-6, 3- (nitrilomethylene bridging) imidazo [5,1-f ] [1,4,7,10] benzoxatridecyl-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-3, 6- (nitrilomethylenebridging) pyrrolo [2,1-f ] [1,4,7,10] benzoxatridecyl-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-3, 6- (nitrilomethylene bridging) imidazo [2,1-f ] [1,4,7,10] benzoxatridecyl-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-3, 6-ethenyl [1,2,4] triazolo [3,4-f ] [1,4,7,8,10] benzoxatetraazatridecyl-17 (14H) -one; 9-chloro-10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-6, 3- (nitrilomethylenebridging) [1,2,4] triazolo [3,4-f ] [1,4,7,10] benzoxatridecyl-17 (14H) -one; 8-chloro-9-fluoro-6-methyl-6,7,14,15-tetrahydro-2H-3, 5- (nitrilomethylene bridging) pyrrolo [3,4-f ] [1,4,8,10] benzoxatrinexazatridec-16 (13H) -one; 8-chloro-9-fluoro-6-methyl-6, 7,14, 15-tetrahydro-2H-3, 5- (nitrilomethylenebridging) pyrazolo [3,4-f ] [1,4,8,10] benzoxatrinazatridec-16 (13H) -one; 8-chloro-9-fluoro-6-methyl-6, 7,14, 15-tetrahydro-2H-3, 5- (nitrilomethylenebridging) pyrazolo [3,4-f ] [1,4,10] benzoxadiazidenyl ring-16 (13H) -one; 12-chloro-11-fluoro-5, 14-dimethyl-6, 7,13, 14-tetrahydro-2H-1, 15- (nitrilomethylenebridging) pyrrolo [3,4-f ] [1,4,10] benzoxadiazatridec-4 (5H) -one; (8R) -10-fluoro-8, 16-dimethyl-15, 16-dihydro-8H-3, 6-ethenyl-bridged imidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; 10-fluoro-8, 16-dimethyl-15, 16-dihydro-8H-3, 6-vinylimidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; (7R) -9-fluoro-7, 15-dimethyl-14, 15-dihydro-2H, 7H-3,5- (nitrilomethylenebridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine slow-release-16 (13H) -one; and 9-fluoro-7, 15-dimethyl-14, 15-dihydro-2H, 7H-3,5- (nitrilomethylene bridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine slow-form-16 (13H) -one; or a pharmaceutically acceptable salt thereof.
In other embodiments, the compound of formula (I) or (I-a) is selected from the group consisting of: 12-chloro-11-fluoro-14-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-3, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 10-fluoro-8-methyl-15, 16-dihydro-8H-3, 6-ethenylimidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; 10-fluoro-7-methyl-7, 8,15, 16-tetrahydro-3, 6-ethenylimidazo [5,1-f ] [1,4,7,8,10] benzoxatetrazethazacyclol-17 (14H) -one; 14-ethyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-14-propyl-6,7,13,14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-14- (prop-2-yl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 14-cyclopropyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-14- (2-hydroxyethyl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-6, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 14-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-13-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (13R) -11-fluoro-13-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 12-chloro-11-fluoro-13-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-14-methyl-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl ring-7-carboxamide; 11-fluoro-7- (hydroxymethyl) -14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-13-methyl-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecane-7-carboxamide; 11-fluoro-7- (hydroxymethyl) -13-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-7-carboxamide; 11-fluoro-7- (hydroxymethyl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl ring-13-carboxylic acid methyl ester; 11-fluoro-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-13-carboxamide; 11-fluoro-14-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] pyrido [3,2-l ] [1,4,8,10] oxa-triazatridean-4 (5H) -one; 11-fluoro-13-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] pyrido [3,2-l ] [1,4,8,10] oxatriazatridean-4 (5H) -one; 11-fluoro-13- (prop-2-yl) -6,7,13, 14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] pyrido [3,2-l ] [1,4,8,10] oxa-triazatricyclo-4 (5H) -one; 13-cyclopropyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f ] pyrido [3,2-l ] [1,4,8,10] oxa-tris-azacyclohex-4 (5H) -one; 13-cyclopropyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-13- (propan-2-yl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; 11-fluoro-6, 7-dihydro-13H-1, 15-ethenylpolypyrazolo [4,3-f ] [1,10,4,8] benzoxathiadiazepin-4 (5H) -one; 11-fluoro-6, 7-dihydro-13H-1, 15-ethenylpolypyrazolo [4,3-f ] [1,10,4,8] benzoxathiadiazepin-4 (5H) -one 14, 14-dioxide; 6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [10,1,4,8] benzoxathiadiazatrizepin-4 (5H) -one; 14-methyl-6,7,13,14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzothiepin-4 (5H) -one; 13-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzothiepin-4 (5H) -one; 11-fluoro-6, 7-dihydro-5H-1, 15-ethenylpyrazolo [3,4-e ] [11,1,2,4,8] benzoxathiatridecyl-4 (14H) -one 13, 13-dioxide; 11-fluoro-14-methyl-6, 7-dihydro-5H-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [11,1,2,4,8] benzoxathiotrine-4 (14H) -one 13, 13-dioxide; 12-fluoro-15-methyl-5, 6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g ] [1,5,9,11] benzoxatrinazatetradec-4-one; 12-fluoro-14-methyl-5,6,7,8,14,15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g ] [1,5,9,11] benzoxatrinazatetradec-4-one; (14R) -12-fluoro-14-methyl-5, 6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g ] [1,5,9,11] benzoxathizatetradecan-4-one; 11-fluoro-7, 14-dimethyl-4, 5,6,7,13, 14-hexahydro-8H-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [2,4,10] benzotriazepin-8-one; 11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [7,2,4,10] benzoxatridecyl-8 (5H) -one; 11-fluoro-7, 14-dimethyl-4, 5,6,7,13, 14-hexahydro-8H-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [2,4,7,10] benzotetraazatridecyl ring-8-one; 11-fluoro-4,7,14-trimethyl-4,5,6,7,13,14-hexahydro-8H-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [2,4,7,10] benzotetrazetadienyl-8-one; 11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [7,2,4,10] benzothiepitridecyl-8 (5H) -one; 11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [3,4-e ] [7,2,4,10] benzothiepin-8 (5H) -one 4, 4-dioxide; and 12-fluoro-8, 15-dimethyl-5, 6,7,8,14, 15-hexahydro-9H-1, 16-ethenylpyrazolo [3,4-e ] [7,2,4,8,11] benzothiepin-9-one 4, 4-dioxide; or a pharmaceutically acceptable salt thereof.
In other embodiments, the compound of formula (I) or (I-a) is selected from the group consisting of: 11-chloro-13-methyl-6,7,13,14-tetrahydro-1,15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 13-Ethyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 13-cyclobutyl-11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f ]][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 11-fluoro-14-methyl (6, 7- 2 H 4 ) 6,7,13,14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 11-fluoro-13-phenyl-6, 7,13, 14-tetrahydro-1, 15-ethaneAlkenyl-bridged pyrazolo [4, 3-f)][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 13- (cyclopropylmethyl) -11-fluoro-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (7R, 14R) -12-fluoro-7-hydroxy-14-methyl-5, 6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-pyrazolo [4,3-g][1,5,9,11]Benzoxatritetraazatetradecachene-4-one; (7S,14R) -12-fluoro-7-hydroxy-14-methyl-5,6,7,8,14,15-hexahydro-4H-1,16-ethenylpyrazolo [4,3-g][1,5,9,11]Benzoxatritetraazatetradecachene-4-one; (7R, 13R) -11-fluoro-7, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4, 3-f)][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (7S,13R) -11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (7R) -11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4, 3-f)][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (6R) -11-fluoro-6, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged-pyrazolo [4,3-f ]][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 12-fluoro-7-hydroxy-15-methyl-5, 6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g][1,5,9,11]Benzoxatritetraazan-4-one; (7S) -11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged-pyrazolo [4,3-f ]][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 11-fluoro-13- (hydroxymethyl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 12-fluoro-14- (hydroxymethyl) -5,6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-pyrazolo [4,3-g][1,5,9,11]Benzoxatritetraazatetradecachene-4-one; 11-fluoro-13, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 11-fluoro-14- (2-hydroxy-2-methylpropyl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 12-fluoro-5,6,7,8,14,15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g][1,5,9]Benzoxadiazepin-tetradecyl-4-one; 11-fluoro-14-methyl-6,7,13,14-tetrahydro-1,15-ethenyl-pyrazolo [4,3-f][1,4,8,10]BenzothiatriazenesHeterotridecyl core-4 (5H) -one; 11-fluoro-14- (1-methylpyrrolidin-3-yl) -6,7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; 11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzothiepitrithiazatridecyl-4 (5H) -one 8-oxide; 11-fluoro-14-methyl-6,7,13,14-tetrahydro-1,15-ethenyl-pyrazolo [4,3-f][1,4,8,10]Benzothiepitridecane-4 (5H) -one 8, 8-dioxide; (7S) -11-fluoro-7-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8]Benzoxadiazidexas-4 (5H) -one; (6S, 13R) -11-fluoro-6, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (6R, 13R) -11-fluoro-6, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; (7S,13S) -11-fluoro-13- (hydroxymethyl) -7-methyl-6,7,13,14-tetrahydro-1,15-ethenyl-pyrazolo [4,3-f][1,4,8,10]Benzoxatridecyl-4 (5H) -one; and 11-fluoro-6, 7-dihydro-13H-1, 15-ethenyl-pyrazolo [4,3-f ]][1,10,4,8]Benzoxathiadiazaspiro-4 (5H) -one; or a pharmaceutically acceptable salt thereof.
In other embodiments, the compound of formula (I) or (I-a) is selected from the group consisting of: (13R) -5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (13R) -11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenylpolypyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -12-chloro-11-fluoro-5- (2-hydroxyethyl) -13-methyl-6, 7-dihydro-13H-1, 15-ethenylpolypyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; 2- [ (13R) -12-chloro-11-fluoro-13-methyl-4-oxo-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-5 (4H) -yl ] acetamide; (13R) -12-chloro-11-fluoro-13-methyl-5- (pyrrolidin-2-ylmethyl) -6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -12-chloro-11-fluoro-7- (hydroxymethyl) -5, 13-dimethyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13S) -11-fluoro-13- (fluoromethyl) -5-methyl-6, 7-dihydro-13H-1, 15-ethenylpyrazolo [4,3-f ] [1,10,4,8] benzodiazepine ring-4 (5H) -one; (13R) -13-cyclopropyl-11-fluoro-5-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (13R) -11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,10,4,8] benzodiazepine slow-4 (5H) -one; (13R) -12-chloro-11-fluoro-13-methyl-6, 7-dihydro-13H-1, 15-ethenylpyrazolo [4,3-f ] [1,10,4,8] benzodiazepine-4 (5H) -one; (8R) -9-chloro-10-fluoro-8-methyl-15, 16-dihydro-8H-3, 6-ethenylimidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; (7R) -8-chloro-9-fluoro-7-methyl-14, 15-dihydro-2h, 7H-3,5- (nitrilomethylene bridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine cyclol-16 (13H) -one; (5R) -3-fluoro-5-methyl-14, 15-dihydro-5H, 10H-9,7- (nitrilomethylenebridging) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,10,3,7] dioxadiazepin-12 (13H) -one; (5R) -3-fluoro-5, 16-dimethyl-13, 14,15, 16-tetrahydro-5H-9, 7- (nitrilomethylenebridging) pyrido [2,3-k ] pyrrolo [3,4-d ] [1,3,7,10] oxa-triazatridean-12 (10H) -one; (13R) -12-chloro-11-fluoro-5, 13-dimethyl-6, 7-dihydro-2H, 13H-1,15- (nitrilomethylenebridge) pyrrolo [3,4-f ] [1,10,4] benzodioxazepin-4 (5H) -one; (7R) -8-chloro-9-fluoro-7, 15-dimethyl-14, 15-dihydro-2H, 7H-3,5- (nitrilomethylene bridging) pyrazolo [3,4-f ] [1,10,4] benzodioxazepin-16 (13H) -one; (13R) -12-chloro-11-fluoro-13, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecane-4 (5H) -one; (8R) -10-fluoro-8, 16-dimethyl-15, 16-dihydro-8H-3, 6-ethenyl-bridged imidazo [5,1-f ] [1,10,4,7,8] benzodioxatridecyl-17 (14H) -one; (7R) -9-fluoro-7, 15-dimethyl-14, 15-dihydro-2H, 7H-3,5- (nitrilomethylenebridging) pyrrolo [3,4-f ] [1,10,4,8] benzodiazepine slow-release-16 (13H) -one; (13R) -11-fluoro-13-methyl-6,7,13,14-tetrahydro-1, 15-ethenylpyrrolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (14R) -12-fluoro-14-methyl-5, 6,7,8,14, 15-hexahydro-4H-1, 16-ethenyl-bridged pyrazolo [4,3-g ] [1,5,9,11] benzoxathizatetradecan-4-one; (7R,14R) -12-fluoro-7-hydroxy-14-methyl-5,6,7,8,14,15-hexahydro-4H-1, 16-ethenylpyrazolo [4,3-g ] [1,5,9,11] benzoxacyclotetradecyl-4-one; (7S,14R) -12-fluoro-7-hydroxy-14-methyl-5,6,7,8,14,15-hexahydro-4H-1, 16-ethenylpyrazolo [4,3-g ] [1,5,9,11] benzoxacyclotetradecyl-4-one; (7R, 13R) -11-fluoro-7, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatrinazatridec-4 (5H) -one; (7S,13R) -11-fluoro-7, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatrinazatridec-4 (5H) -one; (7R) -11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (6R) -11-fluoro-6, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (7S) -11-fluoro-7, 14-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; (7S) -11-fluoro-7-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8] benzoxadiazatrix-4 (5H) -one; (6S, 13R) -11-fluoro-6, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f ] [1,4,8,10] benzoxatrinazatridec-4 (5H) -one; (6R, 13R) -11-fluoro-6, 13-dimethyl-6, 7,13, 14-tetrahydro-1, 15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; and (7S,13S) -11-fluoro-13- (hydroxymethyl) -7-methyl-6,7,13,14-tetrahydro-1,15-ethenylpyrazolo [4,3-f ] [1,4,8,10] benzoxatridecyl-4 (5H) -one; or a pharmaceutically acceptable salt thereof.
The following represent illustrative examples of compounds of formula (I) or (I-A):
Figure GDA0003692253770000421
Figure GDA0003692253770000431
Figure GDA0003692253770000441
Figure GDA0003692253770000451
Figure GDA0003692253770000461
Figure GDA0003692253770000471
Figure GDA0003692253770000481
Figure GDA0003692253770000491
Figure GDA0003692253770000501
and pharmaceutically acceptable salts thereof.
Illustrative examples of compounds of formula (I) or (I-A) are represented below:
Figure GDA0003692253770000502
Figure GDA0003692253770000511
Figure GDA0003692253770000521
Figure GDA0003692253770000531
Figure GDA0003692253770000541
Figure GDA0003692253770000551
Figure GDA0003692253770000561
Figure GDA0003692253770000571
and pharmaceutically acceptable salts thereof.
The following represent illustrative examples of compounds of formula (I) or (I-A):
Figure GDA0003692253770000572
Figure GDA0003692253770000581
Figure GDA0003692253770000591
Figure GDA0003692253770000601
Figure GDA0003692253770000611
and pharmaceutically acceptable salts thereof.
Those skilled in the art will recognize that the categories listed or described herein are not exhaustive and that other categories within the defined term may be selected.
Pharmaceutical composition
For therapeutic purposes, pharmaceutical compositions comprising a compound described herein may further comprise one or more pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients are substances that are non-toxic and otherwise biologically suitable for administration to a subject. The excipients facilitate administration of the compounds described herein and are compatible with the active ingredient. Examples of pharmaceutically acceptable excipients include stabilizers, lubricants, surfactants, diluents, antioxidants, binders, colorants, extenders, emulsifiers, or taste modifiers. In a preferred embodiment, the pharmaceutical composition of the invention is a sterile composition. The pharmaceutical compositions can be prepared using mixing techniques known or available to those skilled in the art.
The invention also encompasses sterile compositions, including compositions that comply with national and local regulations governing such compositions.
The pharmaceutical compositions and compounds described herein can be formulated in solutions, emulsions, suspensions, or dispersions in suitable pharmaceutical solvents or carriers, or as pills, tablets, lozenges, suppositories, sachets, dragees, granules, powders, reconstituted powders or capsules along with solid carriers, according to conventional methods known in the art for preparing various dosage forms. The pharmaceutical compositions of the present invention may be administered by a suitable delivery route, for example, by oral, parenteral, rectal, nasal, topical or ocular routes or by inhalation. Preferably, the composition is formulated for intravenous or oral administration.
For oral administration, the compounds of the invention may be provided in solid form (e.g., tablets or capsules), or as solutions, emulsions, or suspensions. To prepare oral compositions, the compounds of the invention can be formulated to obtain, for example, a dose of about 0.1mg to 1g per day, or about 1mg to 50mg per day, or about 50mg to 250mg per day, or about 250mg to 1g per day. Oral tablets may contain the active ingredient in admixture with compatible pharmaceutically acceptable excipients such as diluents, disintegrants, binders, lubricants, sweeteners, flavoring agents, colorants and preservatives. Suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Exemplary liquid oral excipients include ethanol, glycerol, water, and the like. Starch, polyvinylpyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose and alginic acid are exemplary disintegrants. The binder may include starch and gelatin. The lubricant (if present) may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be enteric coated.
Capsules for oral administration include hard and soft gelatin capsules. To prepare hard gelatin capsules, the active ingredient may be mixed with a solid, semi-solid or liquid diluent. Soft gelatin capsules may be prepared by mixing the active ingredient with water, an oil (for example peanut oil or olive oil), liquid paraffin, a mixture of mono-and diglycerides of short chain fatty acids, polyethylene glycol 400 or propylene glycol.
Liquids for oral administration may be in the form of, for example, suspensions, solutions, emulsions or syrups, or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use. The liquid composition may optionally contain: pharmaceutically acceptable excipients, such as suspending agents (e.g., sorbitol, methylcellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel, and the like); non-aqueous vehicles such as oils (e.g., almond oil or fractionated coconut oil), propylene glycol, ethanol, or water; preservatives (e.g., methyl or propyl paraben or sorbic acid); wetting agents, such as lecithin; and, if desired, flavoring or coloring agents.
For parenteral use (including intravenous, intramuscular, intraperitoneal, intranasal or subcutaneous routes), the agents of the invention may be provided in sterile aqueous solutions or suspensions, buffered to the appropriate pH and isotonic or in parenterally acceptable oils. Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. The forms may be presented in unit-dose form (e.g., ampoules or disposable injection devices), in multi-dose form (e.g., vials from which appropriate doses may be removed), or in solid form or preconcentrate useful in the preparation of injectable formulations. An illustrative injection dose ranges from about 1 to 1000 μ g/kg/minute of the mixture of the agent and the pharmaceutical carrier over a period ranging from minutes to days.
For nasal inhalation or oral administration, the pharmaceutical compositions of the present invention can be administered using, for example, a spray formulation also containing a suitable carrier. The compositions of the present invention may be formulated for rectal administration as suppositories.
For topical administration, the compounds of the present invention are preferably formulated as creams or ointments or similar vehicles suitable for topical administration. For topical administration, the compounds of the present invention may be combined with a pharmaceutical carrier at a concentration of about 0.1% to about 10% drug-to-vehicle. Another way of administering the agents of the present invention may be to administer the agents transdermally using a patch formulation.
As used herein, the term "treatment" encompasses both "prophylactic" and "curative" treatment. "prophylactic" treatment is intended to mean delaying the development of a disease, disease symptom, or medical condition, inhibiting an emerging symptom, or reducing the risk of development or recurrence of a disease or symptom. "curative" treatment includes reducing the severity or inhibiting the worsening of an existing disease, symptom, or condition. Thus, treatment includes ameliorating or arresting the worsening of an existing disease symptom, arresting the appearance of other symptoms, ameliorating or preventing the underlying systemic etiology of a symptom, inhibiting a condition or disease (e.g., arresting the development of a condition or disease, relieving a condition or disease, causing regression of a condition or disease, relieving a condition caused by a disease or condition, or terminating the symptoms of a disease or condition).
The term "subject" refers to a mammalian patient, e.g., a human, in need of such treatment.
Exemplary diseases include cancer, pain, neurological diseases, autoimmune diseases and inflammation. Cancers include, for example, lung cancer, colon cancer, breast cancer, prostate cancer, hepatocellular carcinoma, renal cell carcinoma, gastric and esophageal cancers, glioblastoma, head and neck cancer, inflammatory myofibroblast tumors, and anaplastic large cell lymphoma. Pain includes, for example, pain of any origin or etiology, including cancer pain, chemotherapy pain, neuropathic pain, injury pain, or other sources. Autoimmune diseases include, for example, rheumatoid arthritis, sjogren syndrome, type I diabetes, and lupus. Exemplary neurological diseases include Alzheimer's Disease, parkinson's Disease, amyotrophic lateral sclerosis, and Huntington's Disease. Exemplary inflammatory diseases include atherosclerosis, allergy and inflammation due to infection or injury.
In one aspect, the compounds and pharmaceutical compositions of the invention specifically target tyrosine receptor kinases, specifically MET, ALK, AXL, TRK and JAK. Thus, the compounds and pharmaceutical compositions may be used to prevent, reverse, slow or inhibit the activity of one or more of the kinases. In preferred embodiments, the method of treatment targets cancer. In other embodiments, the method is for treating lung cancer or non-small cell lung cancer.
In the inhibition methods of the invention, an "effective amount" is intended to mean an amount effective to inhibit the target protein. The target modulation measurements can be carried out by conventional analytical methods, such as those described below. The modulation can be used in a variety of settings, including in vitro analysis. In the methods, the cell is preferably a cancer cell having aberrant signaling due to MET, ALK, AXL, TRK and/or JAK upregulation.
In the methods of treatment of the present invention, "effective amount" is intended to mean an amount or dose sufficient to produce the desired therapeutic benefit in an individual in need of such treatment. An effective amount or dose of a compound of the invention can be determined by conventional methods (e.g., modeling, dose escalation, or clinical trials) taking into account conventional factors such as mode or route of administration or drug delivery, pharmacokinetics of the agent, severity and course of infection, health and weight of the individual, and the judgment of the treating physician. Exemplary doses are in the range of about 0.1mg to 1g per day, or about 1mg to 50mg per day, or about 50mg to 250mg per day, or about 250mg to 1g per day. The total dose may be a single or divided dose unit (e.g., BID, TID, QID).
After the patient has developed an improvement in the disease, the dosage can be adjusted for prophylactic or maintenance treatment. For example, the dosage or frequency of administration, or both, can be reduced according to the symptoms to an amount that maintains the desired therapeutic or prophylactic effect. Of course, if the symptoms have been alleviated to an appropriate degree, treatment may be discontinued. However, when either symptom recurs, the patient may require intermittent treatment for a long period of time. Patients may also require chronic treatment.
Pharmaceutical combination
The compounds of the invention described herein may be used in combination with one or more other active ingredients in pharmaceutical compositions or methods for the treatment of the diseases and disorders described herein. Other additional active ingredients include other therapeutic agents or agents that mitigate the adverse effects of the therapeutic agent against the intended disease target. The combinations can be used to increase efficacy, ameliorate other disease symptoms, reduce one or more negative effects, or reduce the required dose of a compound of the invention. The additional active ingredients may be formulated as separate pharmaceutical compositions from the compound of the invention or may be included in a single pharmaceutical composition with the compound of the invention. The additional active ingredient may be administered simultaneously with, before or after the administration of the compound of the invention.
Combination agents include those additional active ingredients known or observed to be effective in treating the diseases and conditions described herein, including those effective against another target associated with the disease. For example, the compositions and formulations of the present invention, as well as methods of treatment, may further comprise other drugs or medicaments, such as other active agents that may be used to treat or alleviate the target disease or associated symptoms or conditions. For cancer indications, other such agents include, but are not limited to, kinase inhibitors, such as EGFR inhibitors (e.g., erlotinib, gefitinib); raf inhibitors (e.g., vemurafenib), VEGFR inhibitors (e.g., sunitinib); standard chemotherapeutic agents, such as alkylating agents, antimetabolites, antitumor antibiotics, topoisomerase inhibitors, platinum drugs, mitotic inhibitors, antibodies, hormonal therapy, or corticosteroids. For pain indications, suitable combination medicaments include anti-inflammatory agents, such as NSAIDs. The pharmaceutical compositions of the invention may additionally comprise one or more of the active agents, and the methods of treatment may additionally comprise administering an effective amount of one or more of the active agents.
Chemical synthesis
Exemplary chemical entities useful in the methods of the invention will now be described with reference to the following illustrative synthetic schemes for their general preparation and the following specific examples. One skilled in the art will recognize that to obtain the various compounds herein, the starting materials can be suitably selected such that the final desired substituent will be carried through a reaction scheme with or without appropriate protection to yield the desired product. Alternatively, it may be necessary or desirable to replace the ultimately desired substituent with an appropriate group that can be carried through the reaction scheme and replaced as appropriate with the desired substituent. Moreover, one skilled in the art will recognize that the transformations shown in the following schemes may be performed in any order that is compatible with the functionality of a particular pendant group. Each of the reactions depicted in the general schemes is preferably carried out at a temperature of about 0 ℃ to the reflux temperature of the organic solvent used. Variables are as defined above with reference to formula (I), unless otherwise specified. Isotopically-labeled compounds described herein are prepared according to the procedures described below using appropriately labeled starting materials. Such materials are generally available from commercial suppliers of radiolabelled chemical reagents.
General procedure a:
Figure GDA0003692253770000651
it will be appreciated that compounds of formula a or a-1 can be prepared according to general procedure a using appropriately functionalized starting materials and intermediates.
Step 1. Compound A-1, suitably functionalized (about 1.00 eq.) (wherein R may be added at a suitable temperature (e.g., 0 ℃) A And R B Is a group compatible with the reaction conditions described herein and Nu is a nucleophilic group, e.g., an anion or a group capable of forming a nucleophile, e.g., a halide, to a solution in a reagent capable of promoting the coupling of A-1 to A-2, e.g., an acid (e.g., tfOH (0.6M)) or an alkyl lithium (e.g., n-BuLi)), adding A-2 (where R is R C Is a group compatible with the reaction conditions described herein and X 2 Is, for example, a leaving group) (about 1.00 eq.). The mixture may be stirred at an appropriate temperature (e.g., 60 ℃) until the reaction is complete. The reaction may then be returned to ambient temperature and the reaction mixture may be quenched, neutralized, washed, extracted, dried and/or concentrated under vacuum as desired to obtain a-3.
Step 2. A-3 (wherein R is A 、R B And R C Is compatible with the reaction conditions described herein) (in some exemplary embodiments described herein, A-3 can be a commercially available aldehyde or ketone, or A-3 can be prepared from step 1, about 1.00 eq.) is reacted with a commercially available amine A-4 (where R is C Is a group compatible with the reaction conditions described herein) (about 1.50 eq.) a mixture in a suitable solvent, such as methanol (0.5M), is stirred at a suitable temperature, such as ambient temperature, for a suitable amount of time or until imine formation by TLC or LC-MS is complete. The reducing agent (e.g., naBH) can be added to the reaction solution in portions 4 (about 2.00 eq.)). The mixture may be stirred at an appropriate temperature (e.g., ambient temperature) until TLC or LC-MS indicates completion of the reaction. The reaction may be quenched, washed, extracted, dried and or concentrated under vacuum as desired to provide a-5.
Step 3, prepared or commercially available A-5 (wherein R A 、R B And R C Is a group compatible with the reaction conditions described herein) (about 1 eq.), a commercially available 5-chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (A-6, about 1)eq.) and an appropriate base, such as diisopropylethylamine (about 5 eq.)) in an appropriate solvent, such as butanol (0.4M), may be stirred at an appropriate temperature, such as 110 ℃, for a set length of time or until the reaction appears to be complete. The reaction can be returned to ambient temperature and diluted with water as necessary. The mixture may be extracted, washed, dried, concentrated under reduced pressure and/or purified by chromatography methods as desired to afford a.
In some exemplary methods, general method a can be performed as follows:
Figure GDA0003692253770000661
step 1. A-2 (1.00 eq.) can be added to a solution of A-1 (1.00 eq.) in TfOH (0.6M) at 0 ℃. The mixture may be stirred at 60 ℃ for 4 hours or until the reaction is complete. After cooling to ambient temperature, the reaction mixture can be poured into ice-water (w/w = 1/1) with NaHCO 3 Neutralized to pH about 9 and extracted three times with EtOAc if necessary. The combined organic layers can be washed with brine, optionally over anhydrous Na 2 SO 4 Dried and concentrated to give a-3.
Step 2. A mixture of a-3 (a commercially available aldehyde or ketone, or prepared from step 1, 1.00 eq.) and a commercially available amine a-4 (1.50 eq.) in methanol (0.5M) can be stirred at ambient temperature for 2 hours or by TLC or LC-MS until imine formation is complete. NaBH can be added to the reaction solution in portions 4 (2.00 eq.). The mixture may be stirred at ambient temperature until TLC or LC-MS indicates completion of the reaction. The reaction can be quenched with water and extracted three times with dichloromethane if desired. The combined organic phases can be washed with brine, using anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford a-5.
Step 3. Prepared or commercially available ethyl a-5 (1 eq.), ethyl 5-chloropyrazolo [1,5-a ] pyrimidine-3-carboxylate (a-6,1eq.) and diisopropylethylamine (5 eq.) were heated in butanol (0.4M) at 110 ℃ for 30 minutes or until the reaction was shown to be complete. The reaction can be cooled and diluted with water. The mixture can be extracted four times with dichloromethane (if desired) and the combined extracts can be dried over anhydrous sodium sulfate. After filtration, the mixture can be concentrated under reduced pressure and the residue can be purified via flash chromatography to provide a.
Alternative general procedure a:
Figure GDA0003692253770000671
coupling step 1. A mixture of a suitably functionalized AA-1 (about 1.00 eq.) a suitably functionalized vinyl coupling reagent (about 1.00-1.50 eq.) and a palladium catalyst (about 0.05 eq.) can be heated under suitable reaction conditions under an inert atmosphere to a suitable temperature (e.g., about 90 ℃) for a suitable amount of time until TLC indicates complete consumption of the starting material. The reaction mixture can be poured into H if desired 2 And (4) in O. The mixture can be extracted and the organic phase washed, dried, concentrated, and optionally purified via silica gel column chromatography to obtain AA-2.
Coupling step 2. Compounds of type AA-2 (about 1.00 eq.), 5-chloropyrazolo [1,5-a ] may be added]Pyrimidine-3-carboxylic acid ethyl ester (a-6, about 1.00 eq.) and palladium catalyst are heated under appropriate reaction conditions under an inert atmosphere to an appropriate temperature (e.g., 120 ℃) for an appropriate length of time until TLC indicates complete consumption of the starting material. The reaction mixture can be poured into H if desired 2 And (4) in O. The mixture can be extracted and the organic phase can be washed, dried, concentrated, and optionally purified by silica gel column chromatography to obtain AA-3.
Step 3. Add the appropriate base (molar excess) to a mixture of AA-3 (about 1.00 eq.) and 4-methylbenzenesulfonyl hydrazide (molar excess) in an appropriate solvent at the appropriate temperature under an inert atmosphere. The mixture may be heated to an appropriate temperature (e.g., 65 ℃) and stirred for an appropriate amount of time until TLC indicates completion of the reaction. The mixture can be cooled and concentrated under reduced pressure if desired. The concentrated reaction mixture may be diluted with water and extracted as necessary. The combined organic phases may be washed, dried, filtered, concentrated in vacuo, and purified to obtain AA-4.
General procedure B:
Figure GDA0003692253770000681
step 1. Aldehyde B-1 (about 1.0 eq) can be reacted (where R is A And R B Is a group compatible with the reaction conditions described herein), B-2 (about 1.0 eq) (wherein X is 1 Is a leaving group and PG is a protecting group), a solution of a suitable base (molar excess) and catalyst in a suitable solvent is heated and stirred for a suitable amount of time until the reaction is complete. Additional B-2 can be added and further heated as desired. The mixture can be cooled to ambient temperature and diluted with water as necessary. The mixture may be extracted and the combined extracts may be washed, dried and, if desired, concentrated under reduced pressure. The crude reaction product can be purified via flash chromatography to provide B-3.
Step 2. Aldehyde B-3 (about 1.0 eq) and an appropriately functionalized amine (about 2.0-4.0 eq) in an appropriate solvent (where R is C Is compatible with the reaction conditions described herein) is heated and stirred for an appropriate amount of time. The mixture can be cooled to ambient temperature and a suitable reducing agent (about 1.0 eq) can be added. The mixture can be stirred for an appropriate amount of time and then quenched by the addition of water as needed. The mixture can be extracted with a suitable organic solvent and the combined extracts can be washed, dried and, if desired, concentrated under reduced pressure. The crude reaction product can be purified, if desired, via flash chromatography to provide B-4.
Step 3. Compound B-4 (about 1.0 eq), ethyl 5-chloropyrazolo [1,5-a ] pyrimidine-3-carboxylate (A-6, about 1.0 eq), and a suitable base (molar excess) in a suitable solvent may be heated for a suitable amount of time. The reaction can be cooled and diluted with water if necessary. The mixture can be extracted with a suitable organic solvent, and the combined extracts can be dried and, if desired, concentrated under reduced pressure. The crude reaction product can be purified via flash chromatography to provide B1.
In some exemplary methods, general method B may be performed as follows:
Figure GDA0003692253770000691
step 1. Aldehyde B-1 (about 1.0 eq) can be reacted (where R is A And R B Is a group compatible with the reaction conditions described herein), B-2 (about 1.0 eq) (wherein X is 1 Is a leaving group and PG is a protecting group), potassium carbonate (molar excess) and potassium iodide (catalytic amount) in DMF was heated to 60 ℃ and stirred for about 15 hours. Additional chloride B-2 may be added and further heating at 80 ℃ may be performed as necessary until the reaction appears to be complete. The mixture can be cooled to ambient temperature and diluted by adding water (250 mL) as necessary. The mixture can be extracted with ethyl acetate (3 × 300 mL) and the combined extracts can be washed with water (200 mL) and brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure if necessary. The crude reaction product can be purified via flash chromatography to provide B-3.
Step 2. Aldehyde B-3 (about 1.0 eq) and methylamine (about 2.5 eq) in methanol can be heated to 60 deg.C and stirred for about 1 hour. The mixture can be cooled to ambient temperature and sodium borohydride (about 1.0 eq) can be added. The mixture may be stirred for about 30 minutes and then quenched by the addition of water (200 mL) as needed. The mixture can be extracted with dichloromethane and the combined extracts can be washed with brine (50 mL), dried over sodium sulfate and concentrated if necessary under reduced pressure. The crude reaction product can be purified via flash chromatography to provide B-4.
Step 3. Amine B-4 (about 1.0 eq), ethyl 5-chloropyrazolo [1,5-a ] pyrimidine-3-carboxylate (A-6, about 1.0 eq), and hunig's base (molar excess) in butanol can be heated at 110 ℃ for about 25 minutes. The reaction was cooled and diluted with water (250 mL). The mixture can be extracted with dichloromethane and the combined extracts can be dried over sodium sulfate if desired. The mixture may be concentrated under reduced pressure if necessary. The crude reaction product can be purified via flash chromatography to provide B.
General method C
Figure GDA0003692253770000701
Step 1. May be performed at about 1.0eq. A 、R B 、R C 、R D And R E Is a group compatible with the reaction conditions described herein), X 1 AlkNHPG (about 1.5-2.0 eq.) (wherein X 1 Is a leaving group, alk is a suitably functionalized alkyl group and PG is a protecting group) in a suitable solvent. The mixture may be heated to the appropriate temperature under an inert atmosphere for the appropriate amount of time until LC-MS reveals complete conversion of the starting material to the product. The mixture can be cooled to ambient temperature, diluted with water and extracted with a suitable organic solvent if desired. The combined organic extracts can be washed with water and brine, over Na 2 SO 4 Dried and concentrated if necessary. The resulting residue can be purified via silica gel column chromatography as necessary to obtain C-2.
Step 2, can be performed to C-2 (1 eq.) (wherein R is A 、R C 、R D And R E Is a group compatible with the reaction conditions described herein, alk is an appropriately functionalized alkyl group and PG is a protecting group) in a suitable solvent with the addition of a suitable base (molar excess). The solution may be heated to an appropriate temperature for an appropriate amount of time. The reaction can be neutralized to pH with an appropriate acid<5, and the reaction mixture can be extracted with a suitable organic solvent. The combined organics can be washed and dried if necessary. The crude reaction product mixture can be filtered, concentrated under reduced pressure, and dried under high vacuum if necessary to afford C-3.
Step 3. An appropriate acid (about 4 eq.) may be added to a solution of C-3 (about 1.0 eq.) in an appropriate organic solvent at an appropriate temperature (e.g., 0 ℃). The reaction mixture may be stirred at the appropriate temperature for the appropriate amount of time until the reaction is shown to be complete by LC-MS. The crude product can be filtered, washed, and dried under high vacuum to provide C-4.
Step 4a. A suitable base (molar excess) may be added to C-4 (about 1.0 eq.) in a solvent in a suitable solvent. The solution may be cooled in an ice-water bath and a suitable coupling agent (about 1.5 eq.) may be added to produce an activated ester. The solution can be slowly warmed to ambient temperature and stirred until the starting material is converted to the desired product as evidenced by LC-MS. The mixture can be diluted with water and extracted with a suitable organic solvent if necessary. The combined organic extracts may be washed, dried and, if desired, concentrated under reduced pressure. The resulting residue can be purified by silica gel column chromatography to obtain C.
In some exemplary methods, general method C can be performed as follows:
Figure GDA0003692253770000711
step 1. May be performed at about 1.0eq. A 、R B 、R C 、R D And R E Is a group compatible with the reaction conditions described herein), X 1 AlkNHPG (about 1.5-2.0 eq.) (wherein X 1 Is a leaving group, alk is an appropriately functionalized alkyl group and PG is a protecting group) in DMF (0.5M) adding K 2 CO 3 (about 3.0 eq.). The mixture may be heated at about 80 ℃ for about 2 hours or until complete conversion of the starting material to the product can be indicated by LC-MS. The mixture can be cooled to ambient temperature, diluted with water if necessary and extracted three times with EtOAc if necessary. The combined organic layers can then be washed with water and brine, and then washed with Na 2 SO 4 Dried and concentrated if necessary. The resulting residue can be purified via silica gel column chromatography eluting with EtOAc/hexanes (5-100%, 10 CV) to afford C-2.
Step 2. Lioh.h2o (about 5.0 eq.) may be added to a solution of C-2 (about 1 eq.) in methanol/THF/H2O (3. The solution may be heated at about 70 ℃ for about 2 hours. The reaction may be neutralized to pH at about 0 deg.C with aqueous HCl (2M)<5, and optionally CH 2 Cl 2 And extracting for four times. The combined organic extracts can be washed with brine and, if desired, na 2 SO 4 And (5) drying. The crude product mixture can be filtered, concentrated under reduced pressure, and dried under high vacuum if necessary to afford C-3.
Step 3. C-3 (about 1.0 eq.) may be added to CH at about 0 deg.C 2 Cl 2 (0.25M) was added HCl in dioxane (4M, about 4 eq.). The reaction can be stirred and allowed to warm from 0 ℃ to room temperature for about 27 hours or until the reaction can be shown to be complete by LC-MS. The obtained reactionThe mixture can be filtered and treated with CH 2 Cl 2 Washed and dried under high vacuum if necessary to provide C-4.
And 4a, utilizing HATU to cyclize. DIPEA (about 5.0 eq.) may be added to a solution of C-4 (about 1.0 eq.) in about 10mL DMF (about 0.005M). The solution was cooled in an ice-water bath and HATU (about 1.5 eq.) was added. The solution can be allowed to warm to ambient temperature and stirred until such time as complete conversion of the starting material to the desired product can be demonstrated by LC-MS. The mixture was diluted with water and extracted three times with EtOAc as needed. The combined organic phases can be washed with water and brine, over Na 2 SO 4 Dried and, if necessary, concentrated under reduced pressure. The resulting residue can be purified via silica gel column chromatography (0-5% MeOH/DCM) to obtain C.
Step 4b cyclization with FDPP to DIPEA (ca. 5 eq.) in DMF/CH 2 Cl 2 (3. After complete dissolution of C-4, pentafluorophenyl diphenylphosphinate (FDPP, about 1.05 eq.) was added. The coupling was allowed to stir for 30 minutes or until the reaction was complete as indicated by LC-MS. The reaction solution can utilize CH 2 Cl 2 Diluting with water and Na 2 CO 3 The aqueous solution (2M) and brine were washed three times, optionally over Na 2 SO 4 And (5) drying. After filtration and concentration under reduced pressure, the residue can be chromatographed via a silica gel column using MeOH/CH 2 Cl 2 (0-5%) eluted to purify to provide C.
Examples of the invention
The following examples are provided to illustrate, but not to limit, the invention. One skilled in the art will recognize that the following synthetic reactions and schemes may be modified by selecting appropriate starting materials and reagents to obtain other compounds of formula (I) or (I-a). Bicyclic heteroaromatic compounds with suitable functionality for use in the synthesis process are commercially available.
AbbreviationsThe examples described herein use a variety of materials, including (but not limited to) those described by the following abbreviations known to those skilled in the art:
Figure GDA0003692253770000721
Figure GDA0003692253770000731
example A6
Figure GDA0003692253770000732
Step 1. At 16 ℃ under N 2 To a solution of 5-fluoro-2-hydroxybenzaldehyde (500.00mg, 3.57mmol, 1.00eq.) in MeOH (20.00 mL) was added 1-methylpyrrolidin-3-amine (357.43mg, 3.57mmol, 1.00eq.) in one portion. The mixture was heated at 16 ℃ under N 2 Stirring was continued for 10 hours. Then add NaBH 4 (270.00mg, 7.14mmol, 2.00eq.) and the mixture was heated at 16 ℃ under N 2 Stirring was continued for 6 hours. TLC (DCM: meOH = 15). The reaction mixture was concentrated under reduced pressure to remove MeOH. The residue was diluted with water (50 mL) and extracted with DCM (20 mL. Times.3). The combined organic layers were washed with brine (50 mL) and Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to obtain A6-5 as a yellow solid (350.00mg, 1.56mmol,43.71% yield). 1 HNMR(300MHz,DMSO-d 6 )δ6.94(dd,J=2.7,9.3Hz,1H),6.86(dt,J=3.0,8.6Hz,1H),6.67(dd,J=4.7,8.7Hz,1H),3.71(s,2H),3.24-3.09(m,1H),2.58(dd,J=7.1,8.8Hz,1H),2.48-2.32(m,2H),2.30-2.17(m,4H),2.05-1.82(m,1H),1.60-1.43(m,1H)。
Step 2. At 16 ℃ under N 2 A6-5 (300.00mg, 1.34mmol, 1.00eq.) and 5-chloropyrazolo [1,5-a ] were added in the downward direction]Pyridine-3-carboxylic acid ethyl ester (302.34mg, 1.34mmol, 1.00eq.) to a solution of n-BuOH (40.00 mL) was added DIPEA (1.04g, 8.04mmol, 6.00eq.). The mixture was stirred at 120 ℃ for 2 hours. TLC (PE: etOAc = 1) showed the reaction was complete. The mixture was poured into water (50 mL) and extracted by DCM (50 mL × 3). The mixture was purified by Pre-PLC to obtain A6 formate as a white solid (290.00mg, 701.43umol,52.35% yield).
Example A8
Figure GDA0003692253770000741
To 5-chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (1.25g, 5.54mmol) and a solution of (R) -2- (1-aminoethyl) -4-fluorophenol HCl salt (available from NetChem Inc.) in EtOH (15.83 mL) were added Huynerger's base (3.58g, 27.70mmol) and heated to 70 ℃ for 1.5 hours. The reaction was rotary evaporated to dryness, suspended in water and extracted with DCM (5 × 50 mL). The combined extracts are purified over Na 2 SO 4 Dried and concentrated under reduced pressure. Flash chromatography (ISCO system, silica (40 g), 0-5% methanol in dichloromethane) afforded A8 (1.89g, 5.49mmol,99% yield).
Example A9
Figure GDA0003692253770000751
Step 1. Add propionyl chloride (1.65g, 17.84mmol, 1.00eq.) to a solution of 4-fluorophenol (2.00g, 17.84mmol, 1.00eq.) in TfOH (30.00 mL) at 0 deg.C. The mixture was stirred at 60 ℃ for 4 hours. TLC showed the reaction was complete. The mixture was cooled to 25 ℃, poured into ice-water (w/w = 1/1) (120 mL), and diluted with NaHCO 3 Neutralized to give a pH of about 9, and extracted with EtOAc (120 mL × 3). The combined organic layers were washed with brine (50 mL) over anhydrous Na 2 SO 4 Dried and concentrated to give A9-3 as a colorless oil (1.80g, 10.70mmol,59.98% yield). 1 HNMR(400MHz,CDCl 3 )δ12.09(s,1H),7.45(dd,J=3.0,9.0Hz,1H),7.26-7.20(m,1H),6.97(dd,J=4.5,9.0Hz,1H),3.02(q,J=7.3Hz,2H),1.27(t,J=7.2Hz,3H)。
Step 2. Ammonia gas was bubbled into MeOH (20 mL) at-78 deg.C for 10 min. A9-3 (1.00g, 5.95mmol, 1.00eq.) was added to the solution and stirred at 25 ℃ for 1 hour. Adding Ti (i-PrO) to the reaction mixture 4 (1.63g, 7.14mmol, 1.20eq.), and the mixture was stirred for another 1 hour. Then, add NaBH 4 (449.93mg,11.89mmol, 2.00eq.). The mixture was stirred at 25 ℃ for 12 hours. TLC showed complete consumption of starting material. The residue was poured into water (50 mL) and stirred for 30 minutes. The mixture was filtered and the filtrate was adjusted to pH about 1 with HCl (1M) and extracted with EtOAc (50 mL. Times.2). Sodium bicarbonate was added to the aqueous phase to adjust the pH to about 9 and extracted with DCM (50 mLx 2). The combined organic layers were washed with saturated brine (50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford A9-5 as a yellow solid (310.00mg, 1.83mmol,30.79% yield). 1 HNMR(400MHz,CDCl 3 )δ6.86(dt,J=3.0,8.4Hz,1H),6.79-6.74(m,1H),6.67(dd,J=2.9,8.9Hz,1H),3.98(t,J=7.0Hz,1H),1.92-1.81(m,1H),1.80-1.68(m,1H),0.95(t,J=7.4Hz,3H)。
Step 3. A9-5 is coupled with ethyl 5-chloropyrazolo [1,5-a ] pyrimidine-3-carboxylate in the presence of DIPEA in n-BuOH as described in general procedure A to provide A9.
Examples A13-5: preparation of 2- (1-amino-2-cyclopropylethyl) -4-fluorophenol
Figure GDA0003692253770000761
Step 1. At 25 ℃ under N 2 To a mixture of 2-cyclopropylacetic acid (4.47g, 44.60mmol, 1.00eq.) in DCM (150.00 mL) was added CDI (7.96g, 49.10mmol,1.10 eq.) in one portion. The mixture was stirred at 25 ℃ for 1hr. N-methoxymethyl amine hydrochloride (4.79g, 49.06mmol, 1.10eq.) was then added. And the mixture was stirred at 25 ℃ for another 12 hours. The reaction was quenched with 1N aqueous hydrochloric acid (50 mL) and separated into layers. The aqueous layer was extracted with DCM (30 mL. Times.2). The combined organic layers were washed with 50% saturated aqueous sodium carbonate (50 mL) and saturated brine (30 mL) over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 2-cyclopropyl-N-methoxy-N-methylacetamide as an oil (6.00g, 41.91mmol,93.96% yield). 1 H NMR(400MHz,CDCl 3 )δ3.65(s,1H),3.18(s,1H),2.33(d,J=6.8Hz,2H),1.13-1.02(m,1H),0.57-0.49(m,2H),0.19-0.11(m,2H)。
Step 2, at-78 deg.C under N 2 To a mixture of 2-cyclopropyl-N-methoxy-N-methylacetamide (6.00g, 29.27mmol, 1.00eq.) in THF (100.00 mL) was added N-BuLi (2.5m, 12.88ml, 1.10eq.) dropwise. The mixture was stirred at-78 ℃ for 10 minutes. And the mixture was then treated with 2-bromo-4-fluoro-1-methoxybenzene (4.19g, 29.27mmol, 1.00eq.) in THF (20 mL) for a period of 20 minutes. After stirring for 1 hour at-78 ℃, the mixture was warmed to 25 ℃ and stirred for an additional 1 hour. TLC showed the reaction was complete. The mixture was poured into 10% aqueous HCl (100 mL) and stirred for 10 minutes. The aqueous phase was extracted with ethyl acetate (300 mL. Times.3). The combined organic phases were washed with brine (200 mL) and over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate =50/1,10/1) to obtain 2-cyclopropyl-1- (5-fluoro-2-methoxyphenyl) ethan-1-one as a colorless oil (2.4 g,39.38% yield). 1 H NMR(400MHz,CDCl 3 )δ7.42(dd,J=3.3,8.8Hz,1H),7.15(ddd,J=3.3,7.5,9.0Hz,1H),6.91(dd,J=4.0,9.0Hz,1H),3.91-3.85(m,3H),2.89(d,J=6.8Hz,2H),1.18-1.05(m,1H),0.61-0.50(m,2H),0.20-0.09(m,2H)。
Step 3-N at-78 deg.C 2 To a solution of 2-cyclopropyl-1- (5-fluoro-2-methoxyphenyl) ethan-1-one (500.00mg, 2.40mmol, 1.00eq.) in DCM (10.00 mL) was added BCl dropwise 3 (1M, 3.00mL, 1.25eq.). The mixture was stirred at-78 deg.C for 2hr. TLC showed the reaction was complete. The mixture was warmed to 25 ℃ and poured into ice-water (w/w = 1/1) (10 mL) and stirred for 10 minutes. The aqueous phase was extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated brine (30 mL) and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 2-cyclopropyl-1- (5-fluoro-2-hydroxyphenyl) ethan-1-one (430.00mg, 2.21mmol,92.3% yield) as an oil. 1 H NMR(400MHz,CDCl 3 )δ12.12(s,1H),7.40(dd,J=3.0,8.8Hz,1H),7.24(ddd,J=3.0,7.8,9.0Hz,1H),6.98(dd,J=4.5,9.3Hz,1H),2.88(d,J=6.8Hz,2H),1.23-1.11(m,1H),0.70-0.63(m,2H),0.25(q,J=5.0Hz,2H)。
Step 4, at 25 ℃ under N 2 Downward 2-cyclopropyl-1- (5-fluoro-2-hydroxyphenyl)) Ethyl-1-one (400.00mg, 1.92mmol, 1.00eq.) in MeOH (20.00 mL) was added NH 2 OH.HCl (160.18mg, 2.31mmol, 1.20eq.) and AcONa (189.09mg, 2.31mmol, 1.20eq.) for 12 hours. TLC (petroleum ether/ethyl acetate = 3) showed complete consumption of the starting material. The reaction was quenched with water and then extracted with DCM (30 mL × 3). The combined organic phases were washed with brine (30 mL) and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford the pure product 2-cyclopropyl-1- (5-fluoro-2-hydroxyphenyl) ethan-1-one oxime (400.00mg, 1.79mmol,93.32% yield) as a white solid. The solid was used in the next step without further purification.
Step 5. In N 2 Pd-C (10%, 100 mg) was added to a solution of 2-cyclopropyl-1- (5-fluoro-2-hydroxyphenyl) ethan-1-one oxime (260.00mg, 1.16mmol, 1.00eq.) in MeOH/HCl (10.00mL, 4N). The suspension is degassed under vacuum and H is used 2 Purging is carried out for several times. Mixing the mixture with H 2 (50 psi) at 50 ℃ for 12 hours. LC-MS showed complete consumption of starting material. The reaction mixture was filtered and the filtrate was concentrated to obtain 2- (1-amino-2-cyclopropylethyl) -4-fluorophenol as a white solid (200.00mg, 955.75umol,82.39% yield). 1 H NMR(400MHz,DMSO-d 6 )δ10.44-9.82(m,1H),8.52(br.s.,2H),7.36(dd,J=2.8,9.5Hz,1H),7.07-6.93(m,2H),4.49(d,J=5.5Hz,1H),1.82-1.72(m,2H),0.67-0.55(m,1H),0.43-0.28(m,2H),0.12-0.06(m,1H),(-0.03)-(-0.09)(m,1H)。
Examples A14 to 5: preparation of 2- (amino (phenyl) methyl) -4-fluorophenol
Figure GDA0003692253770000771
Step 1. At 25 ℃ under N 2 NH was added in one portion to a solution of A14-3 (2.00g, 9.25mmol, 1.00eq.) and AcOK (1.10g, 11.20mmol, 1.20eq.) in ethanol (30.00 mL) 2 Oh.hcl (642.80mg, 9.25mmol, 1.00eq.). The mixture was stirred at 25 ℃ for 30 minutes, then heated to 90 ℃ and stirred for 5 hours. TLC showed the reaction was complete. The mixture was concentrated and water (50 mL) was added. With ethyl acetate (5)0 mL. Times.3) the mixture was extracted. The combined organic phases were washed with brine (50 mL) and over anhydrous Na 2 SO 4 Dried, filtered, and concentrated to give (5-fluoro-2-hydroxyphenyl) (phenyl) methanoxime (1.50g, 6.49mmol,70.13% yield) as a yellow solid. 1 HNMR(400MHz,CDCl 3 )δ7.50-7.37(m,5H),7.19-7.07(m,2H),6.71(dd,J=2.9,8.9Hz,1H)。
Step 2. In N at 25 deg.C 2 Next, NH was added in one portion to a mixture of (5-fluoro-2-hydroxyphenyl) (phenyl) methyl ketoxime (900.00mg, 4.18mmol, 1.00eq.) and Zn powder (1.09g, 16.73mmol,4 eq.) in THF (10.00 mL) 4 Cl (2.24g, 41.82mmol, 10.00eq.). The mixture was stirred at 25 ℃ for 30 minutes, then heated to 60 ℃ and stirred for 15 hours. The mixture was concentrated and water (100 mL) was added, followed by extraction with ethyl acetate (50 mL × 3). The combined organic layers were washed with brine and anhydrous Na 2 SO 4 Dried, filtered, and concentrated to give A14-5 as a yellow solid (630.00mg, 2.90mmol,69.38% yield). 1 HNMR(400MHz,CDCl 3 )δ7.42(d,J=7.5Hz,2H),7.33(t,J=7.5Hz,2H),7.27-7.20(m,1H),6.93-6.80(m,2H),6.70(dd,J=4.9,8.7Hz,1H),5.28(s,1H)。
Example A17
Figure GDA0003692253770000781
Step 1. Converting 5- ((2-bromo-5-fluorobenzyl) (methyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (prepared according to general procedure A) (300.00mg, 0.736mmol, 1.00eq.), 2-methylpropane-2-thiol (166.10mg, 1.84mmol, 2.50eq.), pd 2 (dba) 3 (84.72mg, 0.147mmol, 0.20eq.) to a solution in dioxane (8.00 mL) XantPhos (127.87mg, 0.221mmol, 0.30eq.) and K 2 CO 3 (101.81mg, 0.736mmol, 1.00eq.). Degassing the mixture and adding to N 2 Lower to 120 ℃ for 24 hours. TLC (petroleum ether/ethyl acetate = 1) showed complete consumption of starting material. The reaction mixture is poured into H 2 O (20 mL) and extracted with ethyl acetate (50 mLx 3). The organic phase was washed with brine (30 mL) and dried over anhydrousNa 2 SO 4 Dried, concentrated, and purified by silica gel column chromatography (petroleum ether/ethyl acetate = 21 to 1) to obtain 5- ((2- (tert-butylsulfanyl) -5-fluorobenzyl) (methyl) amino) pyrazolo [1, 5-a) as a yellow solid]Pyrimidine-3-carboxylic acid ethyl ester (200.00mg, 0.48mmol,65.18% yield). 1 H NMR(400MHz,CDCl 3 )δ8.34(s,1H),8.29(br.s.,1H),7.60(dd,J=5.9,8.4Hz,1H),7.00(t,J=7.7Hz,1H),6.29(br.s.,2H),5.00(br.s.,2H),4.37(d,J=6.8Hz,2H),3.41(br.s.,3H),1.36-1.20(m,12H)。
Step 2. In N at 0 deg.C 2 Down to 5- ((2- (tert-butylsulfanyl) -5-fluorobenzyl) (methyl) amino) pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxylic acid ethyl ester (300.00mg, 0.720mmol, 1.00eq.) in DCM (8.00 mL) was added BBr dropwise 3 (902.21mg, 3.60mmol, 5.00eq.). The mixture was stirred at 0 ℃ for 2.5 hours. TLC (petroleum ether: ethyl acetate = 1) showed the reaction was complete. The mixture was poured into water (20 mL). The aqueous phase was extracted with dichloromethane (50 mL. Times.3). The combined organic phases were washed with brine (30 mL) and over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by pre-HPLC (column: phenomenex Synergi (Phenomenex Synergi) C18 x 30mm x 4um and conditions: 0.05% by volume HCl-ACN) and lyophilized to obtain A17 HCl salt as a white solid (38.00mg, 0.098mmol,13.61% yield).
Example A18
Figure GDA0003692253770000791
Step 1. 2-bromo-4-fluorophenol (10.00g, 52.36mmol, 1.00eq.), trifluoro (vinyl) -borane potassium salt (9.84g, 66.50mmol, 1.27eq.), cs 2 CO 3 (51.18g, 157.08mmol, 3.00eq.) and Pd (PPh) 3 ) 2 Cl 2 (1.84g, 2.62mmol, 0.05eq.) in THF (90.00 mL) and H 2 The mixture in O (10.00 mL) was degassed and then in N 2 Heated to 90 ℃ for 12 hours. TLC (petroleum ether/ethyl acetate = 10/1) showed complete consumption of starting material. The reaction mixture is poured into H 2 O (100 mL). The mixture is treated with acetic acid BEster (300 mL. Times.3) was extracted. The organic phase was washed with saturated brine (200 mL) and dried over anhydrous Na 2 SO 4 Dried, concentrated, and purified using silica gel column chromatography (eluting with EtOAc/petroleum ether = 1/30) to obtain 4-fluoro-2-vinylphenol as a colorless oil (3.50g, 25.34mmol,48.39% yield). 1 H NMR(400MHz,CDCl 3 )δ7.12(dd,J=3.0,9.5Hz,1H),6.89-6.81(m,1H),6.79-6.73(m,1H),5.75(d,J=17.6Hz,1H),5.64(s,1H),5.39(d,J=11.3Hz,1H)。
Step 2. In N 2 A mixture of 4-fluoro-2-vinylphenol (1.95g, 14.12mmol, 1.00eq.), TBSCl (6.38g, 42.35mmol, 3.00eq.), and 1H-imidazole (5.77g, 84.70mmol, 6.00eq.) -in DCM (20.00 mL) was stirred at 20 ℃ for 5H. TLC (petroleum ether/ethyl acetate = 10). The reaction mixture was poured onto H 2 In O (30 mL). The mixture was extracted with dichloromethane (50 mLx 3). The organic phase was washed with brine (50 mL) and over anhydrous Na 2 SO 4 Dried, concentrated and purified by silica gel column chromatography eluting with petroleum ether to give tributyl (4-fluoro-2-vinylbenzyl) silane (2.30g, 9.11mmol,64.54% yield) as a colorless oil.
Step 3 tributyl (4-fluoro-2-vinylbenzyl) silane (2.30g, 9.11mmol, 1.00eq.) and 5-chloropyrazolo [1,5-a ])]Pyrimidine-3-carboxylic acid ethyl ester (2.06g, 9.11mmol, 1.00eq.), pd (PhCN) 2 Cl 2 A mixture of (174.7mg, 0.455mmol, 0.05eq.) and tri-o-tolylphosphane (277.36mg, 0.911mmol, 0.10eq.), DIPEA (7.07g, 54.68mmol, 6.00eq.) in DMF (25.00 mL) was degassed and then N 2 Lower to 120 ℃ for 24 hours. TLC (petroleum ether/ethyl acetate = 1) showed complete consumption of the starting material. The reaction mixture was poured onto H 2 O (30 mL). The mixture was extracted with ethyl acetate (100 mL. Times.3). The organic phase was washed with saturated brine (30 mL) and dried over anhydrous Na 2 SO 4 Dried, concentrated, and purified by silica gel column chromatography (EtOAc: petroleum ether = 1) to obtain (E) -5- (5-fluoro-2-hydroxystyryl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (1.00g, 2.26mmol,24.86% yield). 1 HNMR(400MHz,CDCl 3 )δ9.29(br.s.,1H),8.50(d,J=7.0Hz,1H),8.28(br.s.,1H),7.84(d,J=16.6Hz,1H),7.20-7.04(m,3H),6.69(d,J=5.8Hz,2H),4.20(q,J=6.9Hz,2H),1.30-1.19(m,3H)。
Step 4. At 20 ℃ under N 2 Downward (E) -5- (5-fluoro-2-hydroxystyryl) pyrazolo [1,5-a ]]To a mixture of pyrimidine-3-carboxylic acid ethyl ester (378.22mg, 1.04mmol, 1.00eq.) and 4-methylbenzenesulfonylhydrazide (3.29g, 17.68mmol, 17.00eq.) in THF (4.00 mL) was added NaOAc (1.71g, 20.80mmol, 20.00eq.) in one portion. The mixture was then heated to 65 ℃ and stirred for 12 hours. TLC showed the reaction was complete. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 45 ℃. Water (100 mL) was added to the residue. The aqueous phase was extracted with ethyl acetate (300 mL. Times.2). The combined organic phases were washed with saturated brine (50 mL) and dried over anhydrous Na 2 SO 4 Dried, filtered, concentrated in vacuo, and purified by pre-HPLC (column: feilomengyntan Max-RP 250 × 50mm × 10um,0.225% FA-ACN) to obtain A18 as a white solid (120.00mg, 0.347mmol,33.42% yield).
Example A20
Figure GDA0003692253770000801
To 4-fluoro-2-methylaminomethyl-phenol (305.2mg, 1.97mmol) and 6-chloro-imidazo [1,2-b ]]To a mixture of pyridazine-3-carboxylic acid ethyl ester (230mg, 1.02mmol) in DMSO (5 mL) was added KF (180mg, 3.01mmol). The reaction mixture was stirred at 120 ℃ under nitrogen for 18 hours. The solution was then cooled to ambient temperature, diluted with water (20 mL) and extracted with EtOAc (3X 50 mL). The combined organic layers were further washed with water (3X 50 mL) and brine (50 mL) and washed with Na 2 SO 4 Dried and concentrated. The residue was then purified by silica gel column eluting with EtOAc/hexanes (0-50%, 10 CV) to give the desired product as a white solid (240mg, 69%).
Example A22
Figure GDA0003692253770000811
A22-1 was synthesized according to general procedure A. To a solution of a22-1 (150mg, 0.387mmol) in ethanol (2 mL) was added 4M HCl in dioxane (2 mL) and the reaction solution was heated at 75 ℃ for 2 hours. The solvent was evaporated and the residue was neutralized with Et3N and on a silica gel cartridge with methanol/CH 2 Cl 2 (0-12.5%) to provide A22 (144mg, 100%).
Example A23
Figure GDA0003692253770000812
Step 1 to (5-fluoro-2-methoxyphenyl) methanethiol (496.1mg, 2.88mmol) and 6-chloro-imidazo [1,2-b ]]DIPEA (1.12g, 8.64mmol) was added to a mixture of pyridazine-3-carboxylic acid ethyl ester (650.0mg, 2.88mmol) and ethanol (14.4 mL). The reaction mixture was stirred at 80 ℃ for 1 hour. The reaction mixture was cooled to ambient temperature, diluted with water (50 mL) and extracted with DCM (3 × 50 mL). The combined extracts are purified over Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO System, silica (120 g) eluting with EtOAc/hexanes (0-50%) to afford A23-2 (560mg, 54% yield). A23-2 precipitated from the column during purification.
Step 2. To a solution of a23-2 (498.7mg, 1.38mmol) in methanol (100 mL) was added 4M HCl in dioxane (10 mL) and the reaction solution was heated at 75 ℃ for 2 hours. The solvent was evaporated and the residue was neutralized with Et3N and on a silica gel cartridge using methanol/CH 2 Cl 2 (0-12.5%) was eluted to provide A23 (470mg, 98%).
A1-A24 are prepared according to general procedure A and the methods described herein.
Figure GDA0003692253770000813
Figure GDA0003692253770000821
Figure GDA0003692253770000831
Figure GDA0003692253770000841
Figure GDA0003692253770000851
Figure GDA0003692253770000861
Figure GDA0003692253770000871
Figure GDA0003692253770000881
Figure GDA0003692253770000891
Example B7
Figure GDA0003692253770000892
Step 1. To a mixture of 1- (5-fluoro-2-hydroxy-phenyl) -ethanone (773mg, 5.0mmol) and (2-chloro-ethyl) -carbamic acid tert-butyl ester (1.80g, 10.0mmol) in DMF (20 mL) were added KI (2.0mg, 0.012mmol) and Cs 2 CO 3 (3.26g, 10.0 mmol). The mixture was stirred at 80 ℃ overnight. The mixture was then cooled to ambient temperature, diluted with EtOAc, and washed with 1N NaOH (5 × 10 mL) until LCMS showed no 1- (5-fluoro-2-hydroxy-phenyl) -ethanone peakAnd (4) stopping. Subjecting the organic layer to Na 2 SO 4 Dried and concentrated. The residue was then purified by silica gel column eluting with EtOAc/hexanes (0-30%, 10 CV) to afford the desired product B7-2 as a yellow solid (1.1g, 73.8%): LC-MS (ESI) M/z 320.3 (M + Na) +
Step 2. To a solution of B7-2 (1.0 g, 3.36mmol) in MeOH (10 mL) was added NaBH portionwise 4 (640mg, 16.8mmol). The mixture was stirred at ambient temperature for 2 hours until no starting material remained by LCMS. The solution was then diluted with water (50 mL) and extracted with DCM (3X 20 mL). The combined DCM layers were washed with Na 2 SO 4 Dried and concentrated. The residue was purified by silica gel column eluting with EtOAc/hexanes (0-50%, 10 CV) to give the desired product B7-3 as a pale yellow solid (0.75g, 75%). LC-MS (ESI) M/z 322.3 (M + Na) +1 H NMR (500 MHz, chloroform-d) δ 7.11 (dd, J =9.2,3.4hz, 1h), 6.89 (ddd, J =9.0,7.9,3.2hz, 1h), 6.77 (dd, J =8.9,4.4hz, 1h), 5.09 (q, J =6.6hz, 1h), 4.92 (d, J =4.4hz, 1h), 4.03 (t, J =5.2hz, 2h), 3.62-3.50 (m, 2H), 1.49 (d, J = 6.hz, 43h), 1.45 (s, 9H).
And step 3: to B7-3 (600mg, 2.0 mmol) and {2- [ 4-fluoro-2- (1-hydroxy-ethyl) -phenoxy ] at-78 deg.C]A solution of-ethyl } -carbamic acid tert-butyl ester (450mg, 2.0 mmol) in dry THF (40.0 mL) was added NaH (60%, 80mg,2.0 mmol) in portions. The suspension was stirred at-78 ℃ for 4 hours and allowed to warm to 0 ℃ and stirred for a further 4 hours. The mixture was then placed in a refrigerator at-20 ℃ overnight. LC-MS showed good conversion to the desired product. The mixture was then quenched with a mixture of ice and 1N HCl and extracted with EtOAc (3X 20 mL). The organic layer was washed with Na 2 SO 4 Dried, concentrated and purified twice to afford the desired product B7 as a yellow solid (240mg, 25%):
B1-B7 were prepared according to general procedure B and the methods described herein.
Figure GDA0003692253770000901
Figure GDA0003692253770000911
Figure GDA0003692253770000921
Examples 2 and 2-1.
Figure GDA0003692253770000922
Synthesis A:
example 2 can be prepared as shown in the following scheme starting with a racemic or enantiomerically enriched starting material:
Figure GDA0003692253770000923
Figure GDA0003692253770000931
step 1. To a mixture of compound 2A (1 eq) and 2B (1.2 eq) in anhydrous DMF (0.2M) is added Cs 2 CO 3 (1.5 eq.) and the reaction was heated in an oil bath at 80 ℃ under nitrogen overnight. The mixture was cooled, poured into water, and extracted three times with EtOAc. The combined organic layers were washed five times with water, washed with brine and over Na 2 SO 4 And (5) drying. After concentration, the residue was purified on flash column eluting with EtOAc/hexanes to provide compound 2C.
Step 2. Add NaH (1.2 eq) to a solution of compound 2C (1 eq) in anhydrous THF (0.2M). The reaction mixture was stirred at ambient temperature for 0.5 hours. Compound 2D was added to the mixture and the reaction was heated under nitrogen at reflux overnight. The reaction was cooled to ambient temperature and diluted with a portion of water (1/3 of the volume of THF) and NaOH (3 equivalents). The mixture was stirred and heated at 70 ℃ for 2 hours or until the ester was completely hydrolyzed to the corresponding acid. After cooling, the organic layer was separated and the aqueous layer was neutralized to pH about 5. The resulting precipitate was filtered, washed three times with water, and dried under vacuum to afford compound 2E, which was used without further purification.
Step 3 to Compound 2E (1 equiv) in CH 2 Cl 2 (0.2M) 4M HCl in dioxane (10 equivalents) was added and the mixture was stirred until complete conversion of compound 2E to compound 2F. The mixture was concentrated and the residue was purified by reverse phase preparative HPLC to provide compound 2F.
Step 4. A solution of compound 2F (1 eq) and DIPEA (10 eq) in DMF (0.2M) was added dropwise to a solution of HATU (1.4 eq) in DMF (0.1M) at 0 ℃. After the addition was complete, the mixture was stirred at 0 ℃ for a further 30 minutes. Water was added and the mixture was extracted three times with EtOAc. The combined organic layers were washed with saturated NaHCO 3 Washed twice, then brine, over Na 2 SO 4 Dried and concentrated. The residue was purified on a silica gel column eluting with EtOAc/hexanes to provide example 2.
Synthesis of B:
examples 2 and 2-1 can also be prepared according to the following scheme using either racemic or enantiomerically enriched starting materials:
Figure GDA0003692253770000941
step 1 compound 2C is reacted with compound 2G under the conditions described in step 2 of synthesis a to provide compound 2H.
Step 2 compound 2H is converted to compound 2I under the conditions described in step 3 of synthesis a.
Step 3 to a solution of Compound 2I (1 equiv.) and DIPEA (2 equiv.) in toluene (0.01M) was added Pd (P-tBu) 3 ) 2 (1 equivalent). The reaction mixture was heated at 100 ℃ under 4 bar CO overnight and then concentrated. The residue was purified on a silica gel column eluting with EtOAc/hexanes to provide example 2.
Examples 10 and 10-1.
Figure GDA0003692253770000942
Examples 10 and 10-1 can be prepared as shown in the following schemes using either racemic or enantiomerically enriched starting materials:
Figure GDA0003692253770000943
Figure GDA0003692253770000951
step 1 compound 10C was prepared from compounds 10A and 10B using the method described in step 1 of synthesis a of example 2.
Step 2 compound 10E was prepared from compounds 10C and 10D using the method described in step 2 of synthesis a of example 2.
Step 3. Compound 10E (1 eq.) and NH 2 -NH 2 A mixture (10 equivalents) in methanol (0.2M) was heated at reflux until compound 10E was completely converted to compound 10F. The mixture was concentrated and the residue was purified in reverse phase preparative HPLC to provide compound 10F.
Step 4 compound 10F was converted to example 10 according to the method described for step 4 of synthesis a of example 2.
Example 11-1
Figure GDA0003692253770000961
Step 1: to a solution of 2-chloro-3-fluoro-6-hydroxy-benzaldehyde (175mg, 1.0 mmol), bis-tos ethylene glycol (740 mg,2.0 mmol) in ACN (5 mL) was added K 2 CO 3 (276mg, 2.0mmol) and KI (2 mg). The mixture was stirred at 120 ℃ for 24 hours. The solid was filtered off and the filtrate was concentratedAnd purified by column chromatography to obtain the desired product 11-1B as a white solid. This material was used directly in the next step.
Step 2: to a solution of 11-1B (373mg, 1mmol) in ACN (5 mL) was added NaN 3 (650mg, 10mmol) and the mixture was stirred at 120 ℃ for 24 hours. The solid was filtered off and the residue was concentrated and purified by column chromatography to obtain 11-1C as a white solid (200mg, 82%). 1 H NMR (500 MHz, chloroform-d) δ 10.49 (d, J =1.1hz, 1h), 7.31 (dd, J =9.2,8.2hz, 1h), 6.88 (dd, J =9.2,3.7hz, 1h), 4.21 (dd, J =5.4,4.5hz, 2h), 3.67 (dd, J =5.4,4.5hz, 2h).
And step 3: to a solution of 11-1C (100mg, 0.41mmol) in dry THF (5 mL) at-78 deg.C was added methylmagnesium bromide (1N in Et) 2 O, 0.82mL, 0.82mmol). The mixture was allowed to warm to room temperature and stirred for 2 hours until TLC showed no starting material present. The solution was then cooled to 0 ℃ and saturated NH 4 The aqueous OAc solution was quenched and extracted with EtOAc (20 mL. Times.3). The combined organics were passed over Na 2 SO 4 Dried and concentrated. The residue 11-1D was used directly in the next step. 1 H NMR (500 MHz, chloroform-d) δ 6.97 (dd, J =9.2,8.3hz, 1h), 6.77 (dd, J =9.1,4.1hz, 1h), 5.27 (q, J =6.7hz, 1h), 4.34-4.29 (m, 1H), 4.22-4.16 (m, 1H), 4.04-3.98 (m, 1H), 3.95-3.88 (m, 2H), 1.51 (d, J =6.7hz, 3h).
And 4, step 4: to 5-chloro-pyrazolo [1,5-a ] at-78 DEG C]To a solution of pyrimidine-3-carboxylic acid ethyl ester (100mg, 0.44mmol) and 11-1D (110mg, 0.41mmol) in anhydrous THF (5.0 mL) was added NaH (60%, 17mg, 0.44mmol). The mixture was allowed to warm to room temperature and stirred for 8 hours until a large amount of the desired product was formed. The mixture was then diluted with water/ice and extracted with DCM (3 × 20 mL). The organic layer was washed with Na 2 SO 4 Dried, concentrated and purified by silica gel column chromatography to obtain 11-1E (20mg, 0.045mmol, 6%) as a yellow liquid, which was used directly in the next step.
And 5: to a solution of 11-1E (20mg, 0.045mmol) in MeOH (1 mL) was added LiOH (1695g, 0.38mmol), followed by 1mL H 2 And (O). The mixture was stirred at 60 ℃ for 4 hours until LCMS and TLC indicated completion of the reaction. Cooling the solutionTo room temperature, partially concentrated and acidified by 1N HCl until pH 2-3. The aqueous mixture was extracted with DCM (3X 10 mL). Subjecting the organic layer to Na 2 SO 4 Dried and concentrated. The residue 11-1F was used directly in the next step.
Step 6: to a solution of 11-1F (20mg, 0.045mmol) in DCM (5 mL) was added PPh 3 (24mg, 0.09mmol). The solution was stirred for 1 hour until TLC showed complete conversion of the starting material to the desired product. The mixture was then used in the next step without further characterization. 11-1G MS ESI + m/z 417.7(M+Na) +
And 7: DIPEA (0.20mL, 1.15mmol) was added to a solution of 11-1G in DMF (10 mL) obtained from the above step. The solution was frozen in a dry ice/acetone bath and HATU (40.0 mg, 0.11mmol) was added. The solution was allowed to warm slowly to room temperature and LCMS showed clean conversion of the starting material to the desired product. The mixture was then diluted with water (50 mL) and extracted with EtOAc (3X 50 mL). The combined organic layers were washed with water (3X 50 mL) and brine (50 mL) and washed with Na 2 SO 4 And (5) drying. The solvent was removed and the resulting residue was purified by silica gel column chromatography (0-5% MeOH/DCM) to afford the desired product as a white solid (2.6 mg,20% yield).
Examples 14 and 14-1.
Figure GDA0003692253770000971
Examples 14 and 14-1 can be prepared according to the following scheme using either racemic or enantiomerically enriched starting materials:
Figure GDA0003692253770000981
step 1. To a mixture of compounds 14A (1 eq) and 14B (1.2 eq) in anhydrous DMF (0.2M) was added Cs 2 CO 3 (1.5 eq.) and the reaction was heated in an oil bath at 80 ℃ under nitrogen overnight. The mixture was cooled, poured into water, and extracted three times with EtOAc. The combined organic layers were washed five times with water, washed with brine and over Na 2 SO 4 And (5) drying. After concentration, the residue was purified on a flash silica column eluting with EtOAc/hexanes to provide 14C.
Step 2 MeMgBr (3 equiv., 3M in diethyl ether) was added to a cooled (-78 ℃ C.) solution of 14C (1 equiv.) in dry THF (0.2M). The reaction was stirred for 2 hours from-78 ℃ to 0 ℃ and saturated NH 4 Aqueous Cl was quenched and then extracted with EtOAc (2 ×). The organics were over MgSO 4 Dried, filtered and concentrated. The residue was purified by column chromatography on silica eluting with EtOAc/hexanes to give 14D.
Step 3. Add NaH (1.2 eq) to a solution of compound 14D (1 eq) in anhydrous THF (0.2M). The reaction mixture was stirred at ambient temperature for 0.5 h. 14E was added to the mixture and the reaction was heated to reflux under nitrogen overnight. The reaction was cooled to ambient temperature and then poured into water. The product was extracted three times with EtOAc. The combined organics were washed with brine, over Na 2 SO 4 Dried and concentrated. The residue was purified by silica gel column eluting with EtOAc/hexanes to provide product 14F.
Step 4 to compound 14F (1 eq) in CH 2 Cl 2 (0.2M) 4M HCl/dioxane (10 equivalents) was added and the mixture was stirred until all 14F was converted to 14G. After concentration, the residue was purified in reverse phase preparative HPLC to provide 14G.
Step 5 Pd (P-t-Bu) was added to a solution of 14G (1 eq) and DIPEA (2 eq) in toluene (0.01M) 3 ) 2 (1 equivalent). The reaction mixture was heated at 100 ℃ overnight under 4 bar CO and then concentrated. The residue was purified on a silica gel column eluting with EtOAc/hexanes to provide 14.
Examples 15 and 15-1.
Figure GDA0003692253770000991
Examples 15 and 15-1 can be prepared according to the following scheme using either racemic or enantiomerically enriched starting materials:
Figure GDA0003692253770000992
step 1. To a suspension of 15A (1.0 equiv.) in THF (0.15M) was added 2.0M aqueous NaOH (3 equiv.). The homogeneous reaction mixture was stirred overnight and then the organics were removed under reduced pressure. The aqueous residue was brought to a pH of about 4 using 1.0M aqueous HCl. The resulting precipitate was collected by filtration and washed with H 2 O rinse to obtain a solid of 15B. The filtrate was extracted with EtOAc (2 ×), and the organics were concentrated under reduced pressure to provide an additional portion of 15B.
Step 2. By adding concentrated H 2 SO 4 (2.3 mL) carefully added to CrO 3 (2.67 g) and then 10mL of H was used 2 Stock solutions (2.67M) of Jones reagent were prepared by O dilution. To a suspension of 15B (1.0 eq) in acetone (0.067M) was slowly added jones reagent (1.2 eq). The reaction mixture was stirred for 15 minutes and then extracted with i-PrOH and filtered by rinsing with acetone via a pad of celite. The filtrate was concentrated to provide 15C, which was used without further purification.
Step 4. Add NaH (60% in mineral oil, 1.5 equiv.) to a solution of 15C (1.0 equiv.) in DMF (0.40M) at 0 ℃. The reaction mixture was stirred at room temperature for 30 minutes and then cooled back to 0 ℃, and 2- (trimethylsilyl) ethoxymethyl chloride (4.3ml, 1.2 eq) was added slowly. The reaction mixture was allowed to warm to room temperature, stirred for 1 hour, and then H was used 2 O was quenched and extracted with EtOAc (3 ×). Using H 2 The combined organics were washed with O (3X) and brine, and then over MgSO 4 Dried and concentrated. The residue was purified by flash silica chromatography eluting with 20-30% EtOAc/hexanes to afford 15D.
Step 5. To a reaction mixture of 14D (1.0 equiv.), copper (I) iodide (0.05 equiv.), 8-hydroxyquinoline (0.1 equiv.), and tripotassium phosphate (2.0 equiv.) in DMF (0.2M) under a nitrogen atmosphere, 15D (1.2 equiv.) was added and the reaction mixture was heated at 120 ℃ for 24 hours. The reaction mixture was cooled to rt and then diluted with EtOAc. The mixture was filtered through a pad of celite and the filtrate was evaporated under vacuum. The crude residue was purified on a silica gel column eluting with EtOAC/hexane to afford 15E.
Step 6. A suspension of 15E (1.0 eq) in 1, 4-dioxane (0.062M) and water (1/3 of THF) at 0 ℃ was treated with sulfamic acid (6.0 eq). A solution of sodium chlorite (1.3 equivalents) and potassium dihydrogen phosphate (12 equivalents) in water (1.2M) was added via a dropping funnel over 20 minutes. After the addition was complete, the ice bath was removed and the reaction mixture was stirred at room temperature for 3 hours. THF was added and the reaction mixture was then stirred at rt for an additional 3 h. The reaction mixture was diluted with water and extracted with EtOAc (2 ×). The combined organic layers were washed with water and brine, and then over Na 2 SO 4 Dried, filtered and concentrated. The residue was triturated with ethyl acetate/hexanes to give 15F.
Step 7. Add compound 15F (1 eq) to CH 2 Cl 2 To the solution in (0.2M) was added 4M HCl/dioxane (10 equivalents) and the mixture was stirred until all 15F was converted to 15G. After concentration, the residue was purified in reverse phase preparative HPLC to provide 15G.
Step 8. A solution of compound 15G (1 eq) and DIPEA (10 eq) in DMF (0.2M) was added dropwise to a solution of HATU (1.4 eq) in DMF (0.1M) at 0 ℃. After the addition was complete, the mixture was stirred at 0 ℃ for a further 30 minutes. Water was added and the mixture was extracted three times with EtOAc. The combined organics were washed with saturated NaHCO 3 Washed twice with brine and over Na 2 SO 4 Dried and evaporated. The residue was purified on a silica gel cartridge eluting with EtOAc/hexanes to provide 15.
Examples 18 and 18-1.
Figure GDA0003692253770001011
Examples 18 and 18-1 can be prepared according to the following scheme using either racemic or mirror image isomer enriched starting materials:
Figure GDA0003692253770001012
step 1. To a reaction mixture of 14D (1.0 eq), 18A (1.2 eq) and copper (I) iodide (0.05 eq) in DMF (0.2M) under nitrogen atmosphere was added NaH (3.0 eq). The reaction mixture was heated at 120 ℃ for 24 h, and then cooled to room temperature and diluted with EtOAc. The mixture was filtered through a pad of celite and the filtrate was evaporated under vacuum. The crude residue was purified on a silica gel column eluting with EtOAc/hexanes to give 18B.
Step 2. To a reaction mixture of 18B (1.0 eq) in DMF (0.2M) was added KOH (2 eq) and I 2 (1.1 equiv.). The reaction mixture was stirred at room temperature for 1 hour and then treated with NaHSO 3 Quenched and extracted with EtOAc. The combined organics were washed with saturated NaHCO 3 Washed twice with brine and over Na 2 SO 4 Dried and evaporated. The residue was purified using a silica gel column eluting with EtOAc/hexanes to provide 18C.
Step 3 to compound 18C (1 equivalent) in CH 2 Cl 2 (0.2M) 4M HCl/dioxane (10 equivalents) was added and the mixture was stirred until all 18C was converted to 18D. After concentration, the residue was purified in reverse phase preparative HPLC to provide 18D.
Step 4 to a solution of 18D (1 equiv.) and DIPEA (2 equiv.) in toluene (0.01M) was added Pd (P-t-Bu) 3 ) 2 (1 equivalent). The reaction mixture was heated at 100 ℃ under 4 bar CO overnight and then concentrated. The residue was purified on a silica gel column eluting with EtOAc/hexanes to provide 18.
Example 20
Figure GDA0003692253770001021
Example 20 was prepared according to the following scheme:
Figure GDA0003692253770001022
step 1. (tert-butyl 2- (4-fluoro-2-formylphenoxy) ethyl) carbamate (20C). A solution of aldehyde 20A (1.5g, 11mmol), chloride 20B (2.1g, 12mmol), potassium carbonate (7.4g, 54mmol) and potassium iodide (36mg, 0.2mmol) in DMF (11 mL) was heated to 60 ℃ and stirred for 15 h. Additional chloride 20B (1.0 g,6 mmol) was added and the reaction was completed by heating at 80 ℃ for another 5 hours. The mixture was cooled to room temperature and diluted by the addition of water (250 mL). The mixture was extracted with ethyl acetate (3 × 300 mL) and the combined extracts were washed with water (200 mL) and brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. Flash chromatography (ISCO system, silica, 0-20% ethyl acetate in hexanes) afforded 20C as a viscous oil (3.0 g, 99%). LRESIMS m/z 306.1[ deg. ] M + Na ]] + ,C 14 H 18 F 1 N 1 Na 1 O 4 Calculated value of 306.1.
Step 2 tert-butyl (2- (4-fluoro-2- ((methylamino) methyl) phenoxy) ethyl) carbamate (20D). Aldehyde 20C (2.5g, 8.8mmol) and methylamine (0.69g, 22mmol) in methanol (88 mL) were heated to 60 ℃ and stirred for 1 hour. The mixture was cooled to room temperature and sodium borohydride (0.33g, 8.8mmol) was added. The mixture was stirred for 30 minutes and then quenched by the addition of water (200 mL). The mixture was extracted with dichloromethane (4 × 100 mL) and the combined extracts were dried with brine (50 mL), sodium sulfate and concentrated under reduced pressure. Flash chromatography (ISCO system, silica, 0-100% (10% methanol in ethyl acetate) in hexanes) afforded the title compound (2.1g, 80%) as a gel. LRESIMS m/z 299.2[ alpha ], [ M ] +H] + ,C 15 H 24 F 1 N 2 O 3 299.2.
Step 3.5- ((2- (2- ((tert-butoxycarbonyl) amino) ethoxy) -5-fluorobenzyl) (methyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (20F). Amine 20D (2.1g, 7.0 mmol), ester 20E (1.59g, 7.0 mmol) and Huniger's base (7.0 mL,5.2g, 40mmol) in butanol (17 mL) were heated at 110 ℃ for 25 minutes. The reaction was cooled and diluted with water (250 mL)And (5) releasing. The mixture was extracted with dichloromethane (4 × 100 mL) and the combined extracts were dried over sodium sulfate. The mixture was concentrated under reduced pressure. Flash chromatography (ISCO system, silica, 20-100% ethyl acetate in hexanes) afforded the title compound (2.1g, 75%) as a solid. LRESIMS m/z 488.3[ deg. ] M + H] + ,C 24 H 31 F 1 N 5 O 5 488.2 calculated for (g).
Step 4.5- ((2- (2- ((tert-butoxycarbonyl) amino) ethoxy) -5-fluorobenzyl) (methyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (20G). Sodium hydroxide (40ml, 2m in water) was added to a stirred solution of ester 20F (2.1g, 4.3mmol) in tetrahydrofuran methanol (3, 2,100ml) at room temperature. The reaction was heated to 60 ℃ and stirred for 6.5 hours. The mixture was cooled to 0 ℃ and acidified with hydrochloric acid (45mL, 2M in water) and then diluted with water (100 mL). The mixture was extracted with ethyl acetate (4 × 150 mL) and the combined extracts were dried with brine (50 mL) and sodium sulfate. The mixture was concentrated under reduced pressure to give the title compound (1.92g, 97%) as a solid. LRESIMS m/z 460.2[ alpha ], [ M ] +H] + ,C 22 H 27 F 1 N 5 O 5 Calculated value of 460.2.
Step 5.5- ((2- (2-aminoethoxy) -5-fluorobenzyl) (methyl) amino) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (20H). Hydrochloric acid (5ml, 4m in dioxane) was added to a stirred solution of carboxylic acid 20G (1.92g, 4.2mmol) in dichloromethane (25 mL) at room temperature. The reaction was stirred for 2 hours, then concentrated under reduced pressure to provide the target compound as a solid. LRESIMS m/z 360.2[ solution of ] M + H] + ,C 17 H 10 F 1 N 5 O 3 Calculated value of 360.2.
Step 6 HATU (1.67g, 4.4 mmol) was added to a stirred solution of carboxylic acid 20H (1.50g, 4.2mmol) and houndsbergine (7.28ml, 5.40g, 41.8mmol) in DMF: dichloromethane (5, 60ml) at-78 ℃ under argon atmosphere. The reaction was allowed to warm slowly to room temperature and stirred for 3 hours, then quenched with water (300 mL). The mixture was extracted with ethyl acetate (3 × 100 mL), then dichloromethane (2 × 100 mL), and the combined extracts were dried with brine (50 mL) and sodium sulfate. The mixture was concentrated under reduced pressure. Fast colorSpectrum (ISCO system, silica, 1-4% methanol in dichloromethane), followed by recrystallization from ethyl acetate/methanol afforded example 20 as a solid (0.98g, 68%,2 steps). LRESIMS m/z 342.2[ solution of ] M + H] + ,C 17 H 17 F 1 N 5 O 2 Calculated value of (2) 342.1; 1 H NMR(500MHz,DMSO-d 6 )δ9.43(dd,J=6.9,2.7Hz,1H),8.76(d,J=7.9Hz,1H),8.10(s,1H),7.19-7.25(m,1H),7.03-7.07(m,2H),6.72(d,J=7.9Hz,1H),5.64(dd,J=14.9,1.5Hz,1H),4.48(dt,J=10.2,4.3Hz,1H),4.04-4.10(m,2H),3.81-3.87(m,1H),3.58(s,3H),3.38-3.46(m,1H)。
alternative synthesis of example 20:
example 20 was also prepared by the following alternative route:
Figure GDA0003692253770001041
step 1.5-oxo-4H-pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (20J). At 20 ℃ under N 2 Cs was added in one portion to a solution of 20I (150.00g, 1.08mmol) and ethyl (E) -3-ethoxyprop-2-enoate (292.16g, 2.03mol) in DMF (3.2L) 2 CO 3 (656.77g, 2.02mol). The mixture was stirred at 110 ℃ for 6 hours. The mixture was cooled to 20 ℃ and filtered through a pad of celite. The filter cake was washed with ethyl acetate (3X 30 mL). Adding the filtrate to H 2 O (2L) and acidified with HOAc to pH =4. The resulting precipitate was filtered to obtain 20J as a white solid (173.00g, 834.98mmol,86.36% yield). 1 H NMR(400MHz,DMSO-d 6 )δ8.54(d,J=7.91Hz,1H),8.12(s,1H),6.13(d,J=7.91Hz,1H),4.27(q,J=7.11Hz,2H),1.28(t,J=7.09Hz,3H)。
Step 2.5-Chloropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (20K). At 20 ℃ under N 2 POCl was added to a mixture of 20J (158.00g, 762.59mmol) in MeCN (1.6L) at below 3 (584.64g, 3.81mol). The mixture was stirred at 100 ℃ for 2 hours. The mixture was cooled to 20 ℃ and poured in portions at 0 ℃ into ice-water (5000 mL) and stirred for 20 minutes. The precipitate was filtered and dried to obtain a white color20K (110.00g, 487.52mmol,63.93% yield) as a solid. 1 H NMR(400MHz,DMSO-d 6 )δ9.33(d,J=7.28Hz,1H),8.66(s,1H),7.41(d,J=7.15Hz,1H),4.31(q,J=7.15Hz,2H),1.32(t,J=7.09Hz,3H)。
Step 3.4-fluoro-2-methylaminomethyl-phenol (20M). At 25 ℃ under N 2 To a 20L solution (5.00g, 35.69mmol, 1.00eq.) in MeOH (50.00 mL) was added aqueous methylamine (8.8mL, 71.38mmol,25%,2.00 eq) in one portion. The mixture was stirred at 25 ℃ for 3 hours, then NaBH was added portionwise 4 (2.70g, 71.38mmol, 2.00eq). And the mixture was stirred at 25 ℃ for another 9 hours. TLC showed the reaction was complete. The mixture was concentrated at 45 ℃ under reduced pressure. The residue was poured into water (50 mL). The aqueous phase was extracted with dichloromethane (3X 200 mL) and the combined organic phases were washed with brine (200 mL) over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 20M as a colorless solid (5.10 g,32.87mmol,92.09% yield). 1 H NMR(400MHz,CDCl 3 )δ6.86(dt,J=3.0,8.7Hz,1H),6.78-6.69(m,2H),3.93(s,2H),2.48(s,3H)。
Step 4.5- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]Pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (A1). DIPEA (159.98g, 1.24mol, 5.70eq.) was added to a suspension of 20M (33.70g, 217.17mmol, 1.00eq.) and 20K (49.00g, 217.17mmol, 1.00eq.) in n-BuOH (740.00 mL). The reaction mixture was stirred at 120 ℃ under nitrogen for 2 hours. TLC showed the reaction was complete. The solution was cooled to 25 ℃, and then the solvent was removed. The residue was diluted with water (500 mL) and extracted with dichloromethane (3X 500 mL). The combined organic extracts were washed with brine (300 mL) over anhydrous Na 2 SO 4 Dried and concentrated under vacuum. The residue was triturated with EtOAc (100 mL) to afford A1 as a white solid (60.00g, 174.25mmol,80.24% yield). 1 H NMR (500 MHz, chloroform-d) δ 9.71 (s, 1H), 8.32 (d, J =7.9hz, 1h), 8.30 (s, 1H), 6.98-6.87 (m, 3H), 6.37 (d, J =7.9hz, 1h), 4.82 (s, 2H), 4.42 (q, J =7.1hz, 2h), 3.21 (s, 3H), 1.39 (t, J =7.1hz, 3h).
Step 5.5- { [2- (2-tert-Butoxycarbonylamino-ethoxy) -5-fluoro-benzyl]-methyl-amino } -pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid ethyl esterAn ester (B1). To a solution of A1 (102.85g, 298.6mmol, 1eq.), (2-chloro-ethyl) -carbamic acid tert-butyl ester (56.33g, 313.5mmol, 1.05eq.) in DMF (854 mL) was added K 2 CO 3 (206.41g, 1493mmol, 5.0eq.). The mixture was heated at 80 ℃ for 20 hours, where the conversion of starting material to product by LC-MS was about 85%. To the reaction flask was added an additional portion of (2-chloro-ethyl) -carbamic acid tert-butyl ester (5.633g, 31.35mmol, 0.1eq.) and K 2 CO 3 (41.282g, 298.6mmol, 1eq.). The reaction was stirred at 80 ℃ for a further 21 hours. The mixture was then cooled to room temperature, quenched with water (1000 mL) and extracted with EtOAc (3 × 900 mL). The combined organic extracts were then washed with water (3X 700 mL) and brine (500 mL) and washed with Na 2 SO 4 Dried and concentrated. The resulting residue was purified by silica gel column eluting with EtOAc/hexanes (0-70%) to give B1 as a white solid (128.74g, 96.7% yield). LC-MS (ESI) M/z 510.1 (M + Na) +1 H NMR (500 MHz, chloroform-d) δ 8.30 (s, 1H), 8.26 (s, 1H), 6.92 (td, J =8.6,3.3hz, 1h), 6.83-6.76 (m, 1H), 6.31 (s, 1H), 4.93 (s, 2H), 4.51-4.44 (m, 1H), 4.36 (q, J =7.2hz, 2h), 4.03 (t, J =4.9hz, 2h), 3.69-3.63 (m, 1H), 3.51 (s, 2H), 3.30 (s, 2H), 1.44 (s, 9H), 1.41-1.35 (t, J =7.2hz, 3h).
Step 6.11-fluoro-14-methyl-6, 7,13, 14-tetrahydro-1, 15-ethenyl-bridged-pyrazolo [4,3-f ]][1,4,8,10]Benzoxatridecyl-4 (5H) -one (20). To a solution of B1 (128.74g, 264.07mmol, 1eq.) in methanol (750 mL) and THF (250 mL) was added H 2 LiOH.H in O (250 mL) 2 O (55.40g, 1320mmol, 5.0eq.). The clear solution was heated at 70 ℃ for 2 hours. The reaction was neutralized to pH at 0 ℃ with aqueous HCl (2M, 250mL)<5, and then with CH 2 Cl 2 (1X 1000mL,3X500 mL) was extracted. The combined organics were washed with brine (300 mL) and Na 2 SO 4 And (5) drying. After filtration, evaporation and drying under high vacuum, a white solid was obtained (126.47g, 275.25mmol,104% yield). Acid (121.30g, 264 mmol) was added to CH at 0 deg.C 2 Cl 2 To the solution in (996 mL) was added HCl in dioxane (4 m, 204ml). Stirring was continued from 0 ℃ to room temperature for 27 h until de-Boc was complete as indicated by LC-MS.The white solid was filtered, washed with DCM (400 mL) and dried under high vacuum to provide a white solid of amine 3HCl salt (123.55 g), which was used without further purification. DIPEA (169.4g, 228mL,1310 mmol) in DMF (3.7L) and CH 2 Cl 2 To the solution in (1.0L) was added an acid amine HCl salt (22.92g, 49.0mmol, 1.00eq.). After complete dissolution of the solid, add to CH 2 Cl 2 Pentafluorophenyl diphenylphosphinate (FDPP) (1.1M, 19.76g,51.44mmol, 1.05eq.). Coupling was completed by LC-MS within 30 minutes and then the second part salt and FDPP were added according to the same procedure as the first part. The addition of salt, followed by FDPP was repeated every 30 minutes and each addition cycle was monitored by LC-MS. All salts (123.55g, 264mmol, 1.00eq) and FDPP (106.44g, 277mmol, 1.05eq.) were added in portions to the reaction flask. The reaction solution was concentrated to a volume of about 500mL and a large amount of precipitate was formed. The solid product 20 was filtered and washed with DMF (50 mL. Times.3). The filtrate was poured into water (2L) and additional product precipitated. The solid product was filtered and washed with water (100 mL. Times.3). The combined solid products were dried and re-dissolved in 10% methanol in dichloromethane (1.5L) and then ethyl acetate (1L) was added. The solution was concentrated to about 500mL and a large amount of white solid was formed. After filtration and drying under high vacuum, compound 20 was obtained as a white solid (74.58g, 83% yield).
Powder X-ray diffraction (PXRD) of example 20.
The sample crystalline polymorph 1 of example 20 was transferred to a zero background plate for PXRD analysis. PXRD data were obtained using a Bruker (Bruker) D8X-ray diffractometer according to the manufacturer's recommended procedure. Parameters for scanning: 2-theta range: 4.5 to 39.1 degrees; step size: 0.02 degree; step time: 1 second; analysis time: 180 seconds.
The measured diffraction peaks typically have an error of ± 0.1 degrees (2 θ).
The results are shown in FIG. 1. The data are summarized in table 1.
TABLE 1
2-theta (degree) d-value Peak intensity (count) Peak intensity (%)
10.68 9.611 31.15 5.2
11.96 8.586 19.11 2.9
15.26 6.737 20.92 4.4
19.64 5.244 27.57 6.4
21.94 4.701 452.41 100
23.96 4.309 91.85 18.2
26.82 3.857 10.92 2.2
Differential Scanning Calorimetry (DSC) of example 20.
The DSC measurement shown in FIG. 2 was carried out using a differential scanning calorimeter of the Seiko SSC/5200 type. 7.92mg of the sample crystalline polymorph 1 of example 20 was equilibrated at 36 ℃ and then ramped up to 380 ℃ at a rate of 10 ℃/min. The sample of example 20, crystalline polymorph 1, showed a melting point of 298.9 ℃.
Example 26
Figure GDA0003692253770001071
Example 26 can be prepared according to the following scheme:
Figure GDA0003692253770001081
step 1. Titanium (IV) isopropoxide (1.3 equivalents) was added to a commercial solution of methylamine in methanol (2m, 3 equivalents), followed by the addition of the starting aldehyde 14C (1.0 equivalent). The reaction mixture was stirred at ambient temperature for 5 hours, after which sodium borohydride (1.0 eq) was added and the resulting mixture was further stirred for a period of 2 hours. The reaction was then quenched by addition of water and the resulting inorganic precipitate was filtered and washed with EtOAc. The organic layer was separated and the aqueous portion was further extracted with EtOAc (× 2). The combined extracts are dried (K) 2 CO 3 ) And concentrated in vacuo to afford 26A.
Step 2. A mixture of compound 26A (1 equiv) and DIPEA (2 equiv) in n-BuOH (0.2M) was heated at 120 ℃ overnight, cooled to ambient temperature, and then concentrated. The residue was purified by silica gel column eluting with EtOAc/hexanes to provide product 26B.
Step 3 to compound 26B (1 eq) in CH 2 Cl 2 (0.2M) 4M HCl/dioxane (10 equivalents) was added and the mixture was stirred until all 26B was converted to 26C, after concentration, the residue was purified in reverse phase preparative HPLC to afford 26C.
Step 4. Addition of Pd (P-t-Bu) to a solution of 26C (1 eq) and DIPEA (2 eq) in toluene (0.01M) 3 ) 2 (1 equivalent). The reaction mixture was heated at 100 ℃ under 4 bar CO overnight and then concentrated. The residue was purified on a silica gel column eluting with EtOAc/hexanes to provide 26.
Examples 37 and 37-1.
Figure GDA0003692253770001091
Examples 37 and 37-1 can be prepared from racemic or mirror image isomer-enriched starting materials according to the following scheme:
Figure GDA0003692253770001092
step 1 compound 37B was prepared from compound 2C and compound 37A using the method described for step 2 of synthesis a of example 2.
Step 2 compound 37C was prepared from compound 37B using the method described in step 3 of synthesis a of example 2.
Step 3. Example 37 was prepared from compound 37C using the method described in step 4 of synthesis a of example 2.
Examples 38 and 38-1.
Figure GDA0003692253770001093
Examples 38 and 38-1 can be prepared from racemic or enantiomerically enriched starting materials according to the following scheme:
Figure GDA0003692253770001101
step 1 compound 38B was prepared from compounds 2C and 38A as described in step 2 of synthesis a of example 2.
Step 2 compound 38C was prepared from compound 38B using the method described in step 3 of synthesis a of example 2.
Step 3. Example 38 was prepared from compound 38C using the method described in step 4 of synthesis B of example 2.
Example 39
Figure GDA0003692253770001102
Example 39 was prepared according to the following scheme:
Figure GDA0003692253770001103
Figure GDA0003692253770001111
step 1.2- (3-chloro-4-fluoro-2-formyl-phenoxy) -ethyl]-tert-butyl carbamate (39B). To a solution of 2-chloro-3-fluoro-6-hydroxy-benzaldehyde (39A, 53mg, 0.3mmol) and (2-chloro-ethyl) -carbamic acid tert-butyl ester (135mg, 0.75mmol) in DMF (5 mL) was added KI (2.0 mg, 0.012mmol) and K 2 CO 3 (105mg, 0.75mmol). The mixture was microwaved at 100 ℃ for 2 hours. The mixture was then diluted with water (20 mL) and extracted with EtOAc (3X 20 mL). The combined organic layers were washed with water (3X 20 mL) and brine (20 mL) and washed with Na 2 SO 4 Dried and concentrated to give 39B. The crude residue was used directly in the next step. LC-MS (ESI) m/z 340.3(M+Na) +
Step 2 { [2- (3-chloro-4-fluoro-2-methylaminomethyl-phenoxy) -ethyl]-tert-butyl carbamate (39C). To a solution of 39B (95.4 mg, 0.3mmol) in MeOH (3 ml) was added methylamine hydrochloride (50.7 mg, 0.75mmol). The mixture was stirred at 60 ℃ for 30 minutes. The solution was then cooled to ambient temperature and NaBH was added 4 (11.1mg, 0.3mmol). The mixture was stirred at ambient temperature for 2 hours. The solution was then diluted with water (50 mL) and extracted with DCM (3X 20 mL). The combined organic layers were passed over Na 2 SO 4 Dried and concentrated to give 39C. The crude residue was used directly in the next step. LC-MS (ESI) M/z 333.3 (M + H) +
Step 3.5- { [6- (2-tert-Butoxycarbonylamino-ethoxy) -2-chloro-3-fluoro-benzyl]-methyl-amino } -pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid ethyl ester (39D). To a solution of 20K (67.5mg, 0.3mmol) and 39C (99.9mg, 0.3mmol) in n-BuOH (2.0 mL) was added DIEA (1.0 mL). The mixture was heated at 150 ℃ for 2 hours under microwave. The mixture was then diluted with water and extracted with DCM (3 × 20 mL). The organic layer was washed with Na 2 SO 4 Dried, concentrated and chromatographed on a column of purified silica gel to give 17 as a yellow liquid. LC-MS (ESI) M/z 522.5 (M + H) +
Step 4.5- { [6- (2-tert-Butoxycarbonylamino-ethoxy) -2-chloro-3-fluoro-benzyl]-methyl-amino } -pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid (39E). To a solution of 39D (40mg, 0.0776mmol) in MeOH (1 mL) was added LiOH (1695 mg, 0.38mmol) and H 2 O (1 mL). The mixture was stirred at 60 ℃ for 4 hours. The solution was cooled to ambient temperature, partially concentrated and acidified by aqueous HCl (1N) until pH 2-3. The aqueous mixture was extracted with DCM (3X 10 mL). Subjecting the organic layer to Na 2 SO 4 Dried and concentrated to give 39E. The crude residue was used directly in the next step. LC-MS (ESI) M/z 494.3 (M + H) +
Step 5.5- { [6- (2-amino-ethoxy) -2-chloro-3-fluoro-benzyl]-methyl-amino } -pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid (39F). To a solution of 39E (40mg, 0.0776mmol) in DCM (2 mL) was added TFA (0.4 mL). The solution was stirred for 1 hour. Removing the solvent under rotary evaporationAnd (4) an agent. The residue was redissolved with DCM and re-concentrated (3 ×) to give 39F as a foamy solid. LC-MS (ESI) M/z 393.5 (M + H) +
Step 6. DIEA (0.20mL, 1.15mmol) was added to a solution of 39F (36mg, 0.078mmol) in 10mL of DCM. The solution was frozen in a dry ice/acetone bath and HATU (40.0 mg, 0.11mmol) was added. The solution was slowly warmed to ambient temperature. The mixture was diluted with water (50 mL) and extracted with EtOAc (3X 50 mL). The combined organic layers were washed with water (3X 50 mL) and brine (50 mL) and washed with Na 2 SO 4 Dried and concentrated. The resulting residue was purified by silica gel column (0-5% MeOH/DCM) to give example 39 (6.2 mg, 23.4%) as a white solid. LC-MS (ESI) M/z376.5 (M + H) +1 H NMR (500 MHz, chloroform-d) δ 9.51 (s, 1H), 8.40-8.33 (m, 2H), 7.03 (ddd, J =8.9,8.0,0.7hz, 1h), 6.78 (dd, J =9.3,4.2hz, 1h), 6.40 (d, J =7.9hz, 1h), 5.97 (dd, J =15.0,2.1hz, 1h), 4.49-4.43 (m, 1H), 4.31 (ddd, J =10.9,6.4,4.5hz, 1h), 4.12-4.03 (m, 1H), 3.91 (d, J =14.9hz, 1h), 3.72-3.63 (m, 1H), 3.56 (s, 3H).
Example 40
Figure GDA0003692253770001121
Example 40 was prepared as shown in the following scheme:
Figure GDA0003692253770001131
step 1.5- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]-2-methyl-pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (40B). To a solution of 19A (75mg, 0.14mmol) in methanol (2 mL) was added LiOH (60mg, 1.4mmol) and H 2 O (2 mL). The mixture was stirred at 60 ℃ for 4 hours. The solution was cooled to ambient temperature, partially concentrated and acidified by aqueous HCl (1N) until pH 2-3. The resulting suspension was extracted with EtOAc (3X 20 mL). Subjecting the organic layer to Na 2 SO 4 Dried and concentrated to give 40A. LC-MS (ESI) M/z 331.6 (M + H) +
Step 2.5- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]-2-methyl-pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (2-hydroxy-ethyl) -amide (40B). DIEA (0.20mL, 1.15mmol) and HATU (380.0mg, 1.0mmol) were added to a solution of 40A (140mg, 0.42mmol) and 2-amino-ethanol (244mg, 4 mmol) in DCM (5 mL) at 0 ℃. The solution was slowly warmed to ambient temperature. The mixture was then diluted with water (25 mL) and extracted with EtOAc (3X 25 mL). The combined organic layers were washed with HCl (1N, 3X 20 mL) and brine (50 mL) over Na 2 SO 4 Dried and concentrated. The resulting residue was purified by silica gel column eluting with 0-5% MeOH/DCM (10 CV) to give 40B as a white solid (74mg, 47%). LC-MS (ESI) M/z 374.3 (M + H) +
Step 3. Add PPh to a solution of 40B (74mg, 0.2mmol) in THF (3 mL) and DCM (3 mL) at 0 deg.C 3 (131mg, 0.5 mmol) and di-tert-butyl azodicarboxylate (DTAD) (115mg, 0.5 mmol). The mixture was allowed to warm to ambient temperature and stirred for an additional 4 hours. The solvent was removed and the residue was purified by silica gel column eluting with 0-10% MeOH/DCM (10 CV), followed by preparative TLC to give example 40 as a white solid (15 mg). LC-MS (ESI) M/z 356.5 (M + H) +1 H NMR (500 MHz, chloroform-d) δ 8.12 (d, J =7.7hz, 1h), 6.93 (ddd, J =9.0,3.1,0.9hz, 1h), 6.78 (ddd, J =9.0,7.3,3.0hz, 1h), 6.71 (dd, J =9.1,4.5hz, 1h), 6.28 (d, J =7.7hz, 1h), 5.77 (dd, J =15.2,1.7hz, 1h), 4.38-4.33 (m, 1H), 3.98 (s, 1H), 3.91 (d, J =1.4hz, 1h), 3.78 (dd, J =15.1,0.9hz, 1h), 3.45 (s, 3H), 3.43-3.36 (m, 1H), 2.45 (s, 3H).
Example 41
Figure GDA0003692253770001141
Example 41 was prepared using the method shown in the following scheme:
Figure GDA0003692253770001142
step 1: [2- (2-acetyl-4-fluoro-phenoxy) -ethyl]-aminomethylTert-butyl ester (41B). To a mixture of 1- (5-fluoro-2-hydroxy-phenyl) -ethanone (41A, 773mg, 5.0mmol) and (2-chloro-ethyl) -carbamic acid tert-butyl ester (1.80g, 10.0mmol) in DMF (20 mL) was added KI (2.0mg, 0.012mmol) and Cs 2 CO 3 (3.26g, 10.0 mmol). The mixture was stirred at 80 ℃ overnight. The mixture was then cooled to ambient temperature, diluted with EtOAc, and washed with 1n naoh (5 × 10 mL) until LCMS showed no 1- (5-fluoro-2-hydroxy-phenyl) -ethanone peak. Subjecting the organic layer to Na 2 SO 4 Dried and concentrated. The residue was then purified by silica gel column eluting with EtOAc/hexanes (0-30%, 10 CV) to give the desired product 41B as a yellow solid (1.1g, 73.8%). LC-MS (ESI) M/z 320.3 (M + Na) +
Step 2 tert-butyl (2- (4-fluoro-2- (1-hydroxyethyl) phenoxy) ethyl) carbamate (41C). To a solution of 41B (1.0 g, 3.36mmol) in MeOH (10 mL) was added NaBH in portions 4 (640mg, 16.8mmol). The mixture was stirred at ambient temperature for 2 hours. The solution was then diluted with water (50 mL) and extracted with DCM (3X 20 mL). The combined DCM layers were washed with Na 2 SO 4 Dried and concentrated. The residue was purified by silica gel column eluting with EtOAc/hexanes (0-50%, 10 CV) to give the desired product as a pale yellow solid (0.75g, 75%). LC-MS (ESI) M/z 322.3 (M + Na) +1 H NMR (500 MHz, chloroform-d) δ 7.11 (dd, J =9.2,3.4hz, 1h), 6.89 (ddd, J =9.0,7.9,3.2hz, 1h), 6.77 (dd, J =8.9,4.4hz, 1h), 5.09 (q, J =6.6hz, 1h), 4.92 (d, J =4.4hz, 1h), 4.03 (t, J =5.2hz, 2h), 3.62-3.50 (m, 2H), 1.49 (d, J = 6.hz, 43h), 1.45 (s, 9H).
Step 3.6- {1- [2- (2-tert-Butoxycarbonylamino-ethoxy) -5-fluoro-phenyl]-ethoxy } -imidazo [1, 2-b)]Pyridazine-3-carboxylic acid ethyl ester (41D). 41C (600mg, 2.0 mmol) and {2- [ 4-fluoro-2- (1-hydroxy-ethyl) -phenoxy ] at-78 deg.C]To a solution of-ethyl } -carbamic acid tert-butyl ester (450mg, 2.0 mmol) in anhydrous THF (40.0 mL) was added NaH (60%, 80mg,2.0 mmol) in portions. The suspension was stirred at-78 ℃ for 4 hours and allowed to warm to 0 ℃ and stirred for a further 4 hours. The mixture was then placed in a refrigerator at-20 ℃ overnight. The mixture was then quenched with a mixture of ice and 1N HCl and extracted with EtOAc (3 × 20 mL). Organic layerThrough Na 2 SO 4 Dried, concentrated and purified twice to obtain the desired product as a yellow solid (240mg, 25%). LC-MS (ESI) M/z 511.6 (M + Na) +1 H NMR (500 MHz, chloroform-d) δ 8.16 (s, 1H), 7.90 (d, J =9.7hz, 1h), 7.16 (dd, J =9.0,3.2hz, 1h), 0.95 (d, J =9.5hz, 1h), 6.90-6.88 (m, 1H), 6.81-6.78 (m, 1H), 6.68 (q, J =6.2hz, 1h), 5.84-5.68 (m, 1H), 4.38 (q, J =7.2hz, 2h), 4.15-4.09 (m, 2H), 3.60-3.52 (m, 2H), 1.65 (d, J =6.4hz, 3h), 1.38 (d, J =7.2hz, 3h), 1.35 (s, 9H).
Step 4 compound 41D was converted to example 41 using methods similar to those described herein. MS:343.2 (M + H) +1 H NMR (500 MHz, chloroform-d) δ 9.82 (d, J =7.0hz, 1h), 8.27 (s, 1H), 8.09 (d, J =9.5hz, 1h), 7.18 (dd, J =8.9,3.2hz, 1h), 7.01-6.94 (m, 2H), 6.83 (dd, J =9.0,4.3hz, 1h), 6.60-6.53 (m, 1H), 4.63-4.52 (m, 1H), 4.27-4.16 (m, 1H), 4.16-4.04 (m, 1H), 3.70-3.56 (m, 1H), 1.70 (d, J =6.4hz, 3h)
Example 42
Figure GDA0003692253770001151
Example 42 was prepared using the method shown in the following scheme:
Figure GDA0003692253770001152
step 1.6- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]Imidazo [1,2-b ]]Pyridazine-3-carboxylic acid ethyl ester (42B). To 4-fluoro-2-methylaminomethyl-phenol (20L, 305.2mg, 1.97mmol) and 6-chloro-imidazo [1,2-b ]]To a mixture of pyridazine-3-carboxylic acid ethyl ester (42A, 230mg, 1.02mmol) in DMSO (5 mL) was added KF (180mg, 3.01mmol). The reaction mixture was stirred at 120 ℃ under nitrogen for 18 hours. The solution was then cooled to ambient temperature, diluted with water (20 mL) and extracted with EtOAc (3X 50 mL). The combined organic layers were further washed with water (3X 50 mL) and brine (50 mL) and washed with Na 2 SO 4 Dried and concentrated. The residue was then passed through a silica gel column with EtOAc/hexanes (0-50%10 CV) to obtain the desired product as a white solid (240mg, 69%). LC-MS (ESI) M/z 345.2 (M + H) +1 H NMR (500 MHz, chloroform-d) δ 8.61 (s, 1H), 8.17 (s, 1H), 7.91 (d, J =10.0hz, 1h), 7.00-6.86 (m, 4H), 4.78 (s, 2H), 4.47 (qd, J =7.2,0.5hz, 2h), 3.17 (s, 3H), 1.41 (td, J =7.1,0.5hz, 3h).
Step 2.6- { [2- (2-tert-Butoxycarbonylamino-ethoxy) -5-fluoro-benzyl]-methyl-amino } -imidazo [1, 2-b)]Pyridazine-3-carboxylic acid ethyl ester (42C). To 6- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]Imidazo [1,2-b ]]K was added to a solution of pyridazine-3-carboxylic acid ethyl ester (2B, 200mg, 0.58mmol) and (2-chloro-ethyl) -carbamic acid tert-butyl ester (209mg, 1.116mmol) in DMF (5 mL) 2 CO 3 (200mg, 1.45mmol) and KI (2.0 mg, 0.012mmol). The mixture was heated at 90 ℃ for 4 hours under nitrogen. The mixture was then diluted with water (20 mL) and extracted with EtOAc (3X 10 mL). The combined organic layers were then washed with water (3X 5 mL) and brine (2X 5 mL). Subjecting the organic layer to Na 2 SO 4 Dried and concentrated. The resulting residue was purified by passing through a silica gel column eluting with EtOAc/hexanes (0-100%, 10 CV) to afford 42C as a white solid (203mg, 76%). LC-MS (ESI) M/z 510.1 (M + Na) +1 H NMR (500 MHz, chloroform-d) δ (ppm) 8.16 (s, 1H), 7.85 (d, J =9.9hz, 1h), 7.00 (dd, J =8.9,3.2hz, 1h), 6.95-6.87 (m, 2H), 6.80 (dd, J =8.9,4.3hz, 1h), 4.95 (s, 1H), 4.74 (s, 2H), 4.41 (q, J =7.2hz, 2h), 4.04 (t, J =5.2hz, 2h), 3.56-3.50 (m, 2H), 3.26 (s, 3H), 1.43 (s, 9H), 1.40 (t, J =7.2hz, 3h).
Step 3. Compound 42C was converted to example 42 using methods similar to those described herein. MS:342.5 (M + H) +1 H NMR (500 MHz, chloroform-d) δ 10.01 (d, J =6.9hz, 1h), 8.17 (s, 1H), 8.04 (d, J =10.0hz, 1h), 7.07-7.04 (m, 1H), 7.00 (d, J =10.0hz, 1h), 6.96-6.92 (m, 1H), 6.84 (dd, J =9.1,4.5hz, 1h), 5.69 (dd, J =15.8,1.6hz, 1h), 4.55 (dt, J =9.9,3.7, H), 4.20-4.09 (m, ddh), 3.98 (d, J =15.9,1.0hz, 1h), 3.66-3.62 (m, 1H), 3.61 (s, 3H).
Example 51-1
Figure GDA0003692253770001161
Step 1 to a solution of A8 (399.4 mg, 1.16mmol) and tert-butyl (2-chloroethyl) carbamate (260.5 mg, 1.45mmol) in DMF (5.8 mL) was added K 2 CO 3 (801.6mg, 5.80mmol) and heated at 80 ℃ for 6 hours while stirring. The reaction was cooled to ambient temperature and diluted with DCM (3 mL), filtered through a syringe filter and concentrated under reduced pressure. Flash chromatography (ISCO system, silica (12 g), 0-70% ethyl acetate in hexanes) afforded 51-1A (407.4mg, 0.836mmol,72% yield).
Step 2. To a solution of 51-1A (407.4mg, 0.836mmol) in MeOH (6 mL) and THF (4 mL) at ambient temperature was added aqueous LiOH (2M, 4.0 mL). The reaction solution was heated at 70 ℃ for 2 hours. The reaction flask was cooled to ambient temperature, diluted with water and methanol, and then quenched with aqueous HCl (2M, 4 mL) to pH<5. The mixture was extracted with DCM (3X 5 mL) and Na 2 SO 4 Dried, concentrated under reduced pressure, and dried under high vacuum overnight. To a solution of the acid product in DCM (6 mL) was added HCl 4M in 1, 4-dioxane (2.97 mL). The mixture was stirred at room temperature for 3 hours, and then concentrated under reduced pressure and dried under high vacuum. To a solution of de-Boc product and FDPP (352.9mg, 0.918mmol) in DMF (21 mL) at room temperature was added Huynerger's base (539.5mg, 0.327mmol). The mixture was stirred for 2.5 hours and then over 2M Na 2 CO 3 The solution (21 mL) quenched the reaction. The mixture was stirred for 15 min and then extracted with DCM (4 × 10 mL). The combined extracts are purified over Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO system, silica (12 g), 0-11.25% methanol in dichloromethane) to afford 51-1 (164.0 mg,0.480mmol,57.55% yield, three steps).
Example 53
Figure GDA0003692253770001171
Example 53 was prepared using the method shown in the following scheme:
Figure GDA0003692253770001181
step 1.5- [1- (5-fluoro-2-hydroxy-phenyl) -ethylamino]Pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (53A). To 5- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]Pyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid ethyl ester (20M, 300mg, 0.87mmol) in MeOH (5 mL) was added LiOH (420mg, 10mmol), followed by 5mL of H 2 And O. The mixture was stirred at 60 ℃ for 4 hours. The solution was cooled to ambient temperature, partially concentrated and acidified with 1N HCl until pH 2-3. The resulting suspension was extracted with EtOAc (3X 20 mL). Through Na 2 SO 4 The combined organic layers were dried and concentrated. The residue was used directly in the next step. LCMS (ESI) + )m/z 317.4(M+H) +
Step 2.3- ({ 5- [ (5-fluoro-2-hydroxy-benzyl) -methyl-amino]Pyrazolo [1,5-a ]]Pyrimidine-3-carbonyl } -amino) -2-hydroxy-propionic acid methyl ester (53B). To a solution of 53A (80mg, 0.25mmol) and 3-amino-2-hydroxy-propionic acid methyl ester hydrochloride (70mg, 0.5 mmol) in DCM (5 mL) at 0 ℃ was added DIPEA (1.0ml, 5.7 mmol) followed by HATU (140.0mg, 0.5 mmol). The solution was slowly warmed to ambient temperature. The mixture was diluted with water (25 mL) and extracted with EtOAc (3X 25 mL). The combined organic layers were washed with 1N HCl (3X 20 mL) and brine (50 mL) and over Na 2 SO 4 And (5) drying. The solvent was removed and the resulting white solid was used directly in the next step. LC-MS (ESI) + )m/z418.4(M+H) +
Step 3.11-fluoro-14-methyl-4-oxo-4, 5,6,7,13, 14-hexahydro-1, 15-ethenyl-bridged pyrazolo [4,3-f][1,4,8,10]Benzoxatrinazatridecyl ring-7-carboxylic acid methyl ester (53C). To a solution of 53B (83mg, 0.2mmol) in DCM (5 mL) was added PPh 3 (263mg, 1.0mmol), followed by addition of CBr 4 (332mg, 1.0 mmol). The mixture was stirred at ambient temperature overnight. The solvent was removed and the residue was redissolved in DMF (5 mL) followed by addition of K 2 CO 3 (116.8mg, 0.84mmol). The mixture is then stirred at 80 ℃ until the desired product is completely formed asAnd (4) stopping. The mixture was diluted with EtOAc and washed with water. Subjecting the organic layer to Na 2 SO 4 Dried and concentrated. The residue was purified by column on silica gel (0-10%, meOH/DCM) to give 53C as a white solid (40 mg). LC-MS (ESI) + )m/z 400.2(M+H) +
Step 4-addition of NH to 53C (20mg, 0.05mmol) 3 In MeOH (7N, 2mL). The mixture was stirred at 60 ℃ overnight. The solvent was removed and the residue was purified by column on silica gel (0-10%, meOH/DCM) to give example 53 as an off-white solid (8 mg). LC-MS (ESI) + )m/z 385.5(M+H) +1 H NMR (300 MHz, chloroform-d) δ 8.41 (s, 1H), 8.34 (d, J =7.9hz, 1h), 8.17 (s, 1H), 6.99-6.92 (m, 2H), 6.77 (dd, J =6.2,3.5hz, 1h), 6.38 (d, J =7.9hz, 1h), 5.63-5.44 (m, 2H), 5.09 (dd, J =11.0,8.4hz, 1h), 4.38 (dd, J =14.7,11.0hz, 1h), 4.28-4.17 (m, 1H), 4.17-4.07 (m, 2H), 3.22 (s, 3H).
Example 54
Figure GDA0003692253770001191
Example 54 was prepared using the method shown in the following scheme:
Figure GDA0003692253770001192
to a solution of compound 53C (20mg, 0.05mmol) in MeOH (2 mL) was added NaBH portion by portion 4 (19mg, 0.5mmol). The mixture was stirred for 4 hours. The solvent was removed and the residue was purified by column on silica gel (0-10%, meOH/DCM) to give the desired product as a white solid (8 mg). LC-MS (ESI) + )m/z 372.5(M+H) +1 H NMR (300 MHz, chloroform-d) δ 8.39 (s, 1H), 8.32 (d, J =7.9hz, 1h), 7.01-6.85 (m, 3H), 6.35 (d, J =8.0hz, 1h), 5.55-5.43 (m, 1H), 4.92-4.82 (m, 1H), 4.09-3.98 (m, 2H), 3.80-3.70 (m, 3H), 3.23 (s, 3H).
Example 93
Figure GDA0003692253770001193
Figure GDA0003692253770001201
Step 1 to a solution of (R) - (2-hydroxypropyl) carbamic acid tert-butyl ester (1.00g, 5.71mmol) and p-toluenesulfonyl chloride (1.14g, 6.00mmol) in DCM (29 mL) was added triethylamine (1.44g, 14.28mmol) and the mixture was stirred at room temperature for 48 hours. The reaction solution was concentrated under reduced pressure and the residue was purified by flash chromatography (ISCO system, silica (40 g), 0-20% ethyl acetate in hexanes) to afford (R) -4-methylbenzenesulfonic acid 1- ((tert-butoxycarbonyl) amino) propan-2-yl ester (1.12g, 3.40mmol,59.54% yield).
Step 2. To a solution of A8 (100.00mg, 0.290mmol) and (R) -4-methylbenzenesulfonic acid 1- ((tert-butoxycarbonyl) amino) prop-2-yl ester (143.50mg, 0.436 mmol) in DMF (1.45 mL) was added K 2 CO 3 (200.7 mg, 1.45mmol) and heated at 80 ℃ for 16 hours while stirring. The reaction was cooled to ambient temperature and diluted with DCM (3 mL), filtered through a syringe filter and concentrated under reduced pressure. Flash chromatography (ISCO system, silica (12 g), 0-60% ethyl acetate in hexanes) afforded 93A (32.90mg, 0.0656mmol,22.59% yield).
Step 3. To a solution of 93A (32.90mg, 0.0656mmol) in MeOH (3 mL) and THF (2 mL) at ambient temperature was added aqueous LiOH (2M, 2mL). The reaction solution was heated at 70 ℃ for 2 hours. The reaction flask was cooled to ambient temperature, diluted with water and methanol, and then quenched with aqueous HCl (2M, 2mL) to pH<5. The mixture was extracted with DCM (3X 5 mL) and Na 2 SO 4 Dried, concentrated under reduced pressure, and dried under high vacuum overnight. To a solution of the acid product in DCM (4 mL) was added HCl 4M in 1, 4-dioxane (2.0 mL). The mixture was stirred at room temperature for 3 hours, and then concentrated under reduced pressure and dried under high vacuum. To a solution of de-Boc product and FDPP (27.62mg, 0.0719mmol) in DMF (1.6 mL) at room temperature was added Huynerger's base (4)2.23mg, 0.327mmol). The mixture was stirred for 2.5 hours and then over 2M Na 2 CO 3 The solution (2 mL) quenched the reaction. The mixture was stirred for 15 min, then extracted with DCM (4X 10 mL). The combined extracts are purified over Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO system, silica (12 g), 0-10% methanol in dichloromethane) to afford 93 (10.1mg, 0.0284mmol,43.49% yield, three steps).
Examples 104, 106, and 107
Figure GDA0003692253770001211
Step 1. To a solution of A17. HCl (38mg, 0.096 mmol) and tert-butyl (2-chloroethyl) carbamate (12.9mg, 0.072mmol) in DMF (0.5 mL) was added K 2 CO 3 (33.1mg, 0.24mmol) and stirring at 80 ℃ for 1.5 hours while stirring. The reaction was cooled to ambient temperature and diluted with DCM (3 mL), filtered through a syringe filter and concentrated under reduced pressure. Flash chromatography (ISCO system, silica (12 g), 0-60% ethyl acetate in hexanes) afforded 104A (20.8mg, 0.0413mmol,86.3% yield).
Step 2.104 was prepared according to general procedure C from 104A as a white solid.
Step 3. To a solution of 104 (4.6 mg,0.0129 mmol) in DCM (0.3 mL) was added methyl 3-chloroperoxybenzoate (2.2 mg,0.0129 mmol) and the reaction was stirred for 20 min, followed by addition of saturated NaHCO 3 Aqueous (3 mL) and extracted with DCM (4X 4 mL). The combined extracts are purified over Na 2 SO 4 Dried and concentrated under reduced pressure. Flash chromatography (ISCO system, silica (12 g), 0-12.5% methanol in dichloromethane) afforded 106 (0.5mg, 10.4% yield) and 107 (1.7mg, 33.9% yield).
The following examples are prepared using methods similar to those of general methods a, B, and C specifically described herein, as described herein.
Figure GDA0003692253770001212
Figure GDA0003692253770001221
Figure GDA0003692253770001231
Figure GDA0003692253770001241
Figure GDA0003692253770001251
Figure GDA0003692253770001261
Figure GDA0003692253770001271
Figure GDA0003692253770001281
Figure GDA0003692253770001291
Figure GDA0003692253770001301
Figure GDA0003692253770001311
Additional examples are prepared using methods similar to those described above.
Biological example 1: biochemical kinase assay.
MET/ALK/AXL/TRK kinase inhibition can be measured by sequential fluorescence analysis in Munich (Omnia) (Invitrogen Inc.). The reaction was carried out in a volume of 50. Mu.L in a 96-well plate at 30 ℃. The mixture contained 1nM human recombinant target kinase domain, 2. Mu.M phosphate acceptor peptide, test compound (11 doses, 3-fold serial dilutions, 2% DMSO final) or DMSO only, 0.2mM DTT and 10mM MgCl in 20mM Hepes 2 (pH 7.5) and the reaction was initiated by addition of ATP (100. Mu.M final concentration) followed by a20 min pre-incubation. The initial rate of phosphopeptide formation was measured using a taispaniel (Tecan Safire) microplate reader over 20 minutes with the excitation wavelength set at 360nm and the emission wavelength at 485nm. K i Values were calculated by fitting the data to the equation sequence for competitive inhibition using a non-linear regression method (GraphPad Prism, graphPad software, san Diego, CA).
Biological example 2: cellular kinase phosphorylation ELISA assay
The experiments were carried out based on the procedures described in the publications (Christensen J., et al, "PF-2341066 (novel inhibitors of anaplastic lymphoma kinase and c-Met) Cytoreductive antitumor activity in an experimental model of anaplastic large cell lymphoma (cytotoxic activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large cell lymphoma)," molecular cancer therapeutics, 6 (12): 2007): 4-3322.) all experiments were carried out under standard conditions (37 ℃ and 5 CO 2 ℃. (37 ℃ and 5 CO 1.) 2 ) The following procedures were carried out. IC (integrated circuit) 50 Values were calculated by concentration/response curve fitting using a four parameter method based on Microsoft (Microsoft) Excel. Cells were seeded in 96-well plates in medium supplemented with 10% Fetal Bovine Serum (FBS) and transferred to serum-free medium [ with 0.04% Bovine Serum Albumin (BSA) 24 hours later)]In (1). In experiments to study ligand-dependent RTK phosphorylation, the corresponding growth factors were added for up to 20 minutes. Mixing the cells withAfter incubation for 1h with inhibitor and/or with appropriate ligand for a specified time, cells were supplemented with 1mmol/L Na 3 VO 4 Is washed once and protein lysates are produced from the cells. Subsequently, the phosphorylation of the selected protein kinases was assessed by sandwich ELISA method by coating 96-well plates with a specific capture antibody and a detection antibody specific for phosphorylated tyrosine residues. Antibody-coated plates were (a) incubated overnight at 4 ℃ in the presence of protein lysates, (b) washed seven times in Tween (Tween) 20 in 1% PBS, (c) incubated for 30 minutes in horseradish peroxidase-conjugated anti-total phosphotyrosine (PY-20) antibody (1 2 SO 4 Terminating, and (f) measuring absorbance at 450nm using a spectrophotometer. Cell lines for individual kinases include A549 for MET, karpas 299 for ALK, 293-AXL for AXL, PAET RKA for TRKA, and PAE-TRKB for TRKB.
Biological example 3: kinase binding assay.
The kinase binding assay was performed at Discoverx using general KINOMEscan K d Protocol implementation (Fabian, M.A.) et al, "Small molecule kinase interaction maps of clinical kinase inhibitors (A small molecule kinase interaction maps for clinical kinase inhibitors)," Nature Biotechnology (Nat. Biotechnol.) -2005, 23 (3): 329-36). For most assays, kinase-tagged T7 phage strains were prepared in e.coli (e.coli) hosts derived from the BL21 strain. Coli were grown to log phase and infected with T7 phage and incubated with shaking at 32 ℃ until lysis. The lysate was centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently labeled with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase analysis. Blocking of the liganded beads with excess biotin and blocking buffer (SeaBlock (Pierce)), 1% BSA, 0.05% Tween 20.1 mM DTT) to remove unbound ligand and reduce non-specific binding. Binding reactions were pooled by combining kinase, liganded affinity beads and test compound in 1 × binding buffer (20% seablock, 0.17 × PBS, 0.05% tween 20, 6mM DTT). All reactions were performed in polystyrene 96-well plates at a final volume of 0.135 mL. The assay plates were incubated with shaking for 1 hour at room temperature and the affinity beads were washed with wash buffer (1 × PBS, 0.05% tween 20). The beads were then resuspended in elution buffer (1 x PBS, 0.05% tween 20, 0.5 μ M non-biotinylated affinity ligand) and incubated at room temperature for 30 minutes with shaking. The concentration of kinase in the eluate was measured by qPCR. The results of the compounds tested in this assay are presented in table 2. Example 20 also has binding affinity to PLK4 kinase (K) using this method d 2.9nM)。
Table 2.
Figure GDA0003692253770001321
Figure GDA0003692253770001331
Biological example 4: ba/F3 cell proliferation assay.
TRKA Ba/F3 cell proliferation assay was performed by ACD (Advanced Cellular Dynamics). The Ba/F3 cell line was maintained in RPMI-1640 medium containing 10% fetal bovine serum and antibiotics. Cells grown in log phase were harvested and 5,000 cells were distributed in 50 μ L growth medium in each well of 384-well plates. Adding 50 nanoliters of diluted compounds in duplicate to appropriate wells, and humidifying the cells at 37 5% 2 The culture was carried out in an incubator for 48 hours. Viability was determined by adding 15 μ L CellTiter-Glo and measuring luminescence, reported as Relative Light Units (RLU) measured in counts per second. Data (RLU) for each compound was normalized to the mean maximum response obtained in the presence of vehicle (DMSO) only. The data were used to derive% inhibition (100-max)Large response%) and the average of the two data points/concentrations was used to calculate IC via non-linear regression analysis using GraphPad Prism software (GraphPad, inc., san diego, california) 50 Values (concentration that resulted in half maximal inhibition of cell survival). Using this method, example 20 was tested with an IC of 3.0nM 50 Inhibiting cell proliferation of TRKA Ba/F3 cells. Data for the compounds tested in this assay are presented in table 3.
Biological example 5: EML4-ALK Ba/F3 stable cell line production and cell proliferation assays.
The EML4-ALK wild-type gene (variant 1) was synthesized in Kirschrip (GenScript) and cloned into the pCDH-CMV-MCS-EF1-Puro plasmid (systems Biosciences, inc). The Ba/F3-EML4-ALK wild-type cell line was generated by transfecting Ba/F3 cells with a lentivirus containing the EML4-ALK wild-type. Stable cell lines were selected by puromycin treatment followed by IL-3 withdrawal. 5000 cells were seeded overnight in 384-well white plates and then treated with compound. Cell proliferation was measured using the CellTiter-Glo luciferase-based ATP detection assay (Promega) after 48 hours incubation at various concentrations of compound according to the manufacturer's protocol. IC (integrated circuit) 50 The assays were performed using GraphPad Prism software (GraphPad, inc., san diego, california). Data for the compounds tested in this assay are presented in table 3.
Biological example 6: cell proliferation assay.
Colorectal cell line KM12 (with an endogenous TPM3-TRKA fusion gene) cells were cultured in DMEM medium supplemented with 10% fetal bovine serum and 100U/mL penicillin/streptomycin. 5000 cells were seeded in 384-well white plates for 24 hours and then treated with compound. Cell proliferation was measured using CellTiter-Glo luciferase-based ATP detection assay (plomega) after 72 hours of incubation according to the manufacturer's protocol. IC (integrated circuit) 50 The assays were performed using GraphPad Prism software (GraphPad, inc., san diego, california).
The other option is as follows: subjecting colorectal cellsLine KM12 (with endogenous TPM3-TRKA fusion gene) cells were cultured in DMEM medium supplemented with 10% fetal bovine serum and 100U/mL penicillin/streptomycin. Primary thrombocythemia cell line SET-2 cells (with endogenous JAK2V 618F point mutation) or T-cell lymphoma Karpas-299 cell line (with endogenous NPM-ALK fusion gene) were cultured in RPMI medium supplemented with 10% fetal bovine serum and 100U/mL penicillin/streptomycin. 5000 cells were seeded in 384-well white plates for 24 hours and then treated with compound. Cell proliferation was measured using CellTiter-Glo luciferase-based ATP detection assay (promega) after 72 hours of incubation according to the manufacturer's protocol. IC (integrated circuit) 50 The assays were performed using GraphPad Prism software (GraphPad, ltd., san diego, california).
Data for compounds tested in the assay are presented in table 3.
TABLE 3
Figure GDA0003692253770001341
Figure GDA0003692253770001351
Figure GDA0003692253770001361
Biological example 7: cell mechanism of action study-TRKA and downstream signal target phosphorylation assay.
Colorectal cell line KM12 (with endogenous TPM3-TRKA fusion gene) cells were cultured in DMEM medium supplemented with 10% fetal bovine serum and 100U/mL penicillin/streptomycin. One million cells were seeded in 6-well plates for 24 hours and then treated with compound. Cells were washed with 1xPBS and harvested after 5 hours of treatment and plated in RIPA buffer (50mM Tris, pH 7.4,150mM NaCl, riPA buffer supplemented with 10mM EDTA, halt protease and phosphatase inhibitor (Thermo Scientific)),1% NP-40, 0.5% deoxycholate, 0.1% SDS). Protein lysates (20 μ g) were resolved on 4-12% bordetella Bis-Tris pre-gels using MES run buffer (Life Technologies), transferred to nitrocellulose membranes using the Trans-Blot Turbo transfer system (berle) and tested using a targeted phosphorylated TRK a (Cell Signaling Technology), Y496, Y680, Y681, clone C50F3; 1. The antibody was incubated overnight at 4 ℃ with gentle shaking, followed by washing and incubation with the appropriate HRP-conjugated secondary antibody. The membrane was exposed to chemiluminescent substrate for 5 minutes at room temperature (SuperSignal West Femto, seemer tech). Images were obtained using the C-Digit imaging System (LI-COR Biosciences). The relative density of the bands was obtained directly via Image Studio Digits of LICOR. The half Inhibitory Concentration (IC) was calculated using non-linear regression analysis with GraphPad Prism software (GraphPad, inc., san Diego, calif.) 50 ) The value is obtained. Using this method, example 20 was tested with an IC of 1.07nM 50 Inhibition of autophosphorylation of TPM3-TRKA in KM12 cells and IC at 2.80nM and 2.00nM, respectively 50 Inhibiting the phosphorylation of downstream signal targets AKT and ERK.
Biological example 8: caspase activity assay.
KM12 cells were maintained in DMEM medium containing 10% fetal bovine serum and antibiotics. 500,000 cells were seeded in 12-well plates and various concentrations of compounds were introduced for 72 hours. For staurosporine treatment, 500nM STS was added at 60 hours and incubated for 12 hours as a positive control. All cells were collected and washed twice with 1xPBS and then lysed in lysis buffer (20 mM HEPES, 150mM NaCl, 10mM KCl, 5mM EDTA, 1% NPT 40) supplemented with Halt protease and phosphatase inhibitors (Saimer technology). For caspase analysis, approximately 20 μ L (20 μ g) of cell lysate was incubated with 20 μ L of caspase 3glo reagent (Promega), and enzyme activity was measured by luminescence release after incubation for 20 minutes at 37 ℃. For western blotting, cell lysates are boiled and analyzed by SDS-PAGE/immunoblotting using PARP or actin antibodies. Using this method, example 20 induced apoptosis of KM12 cells.

Claims (7)

1. A compound selected from the group consisting of:
Figure FDA0003691104280000011
Figure FDA0003691104280000021
or a salt thereof.
2. A compound selected from the group consisting of:
Figure FDA0003691104280000022
Figure FDA0003691104280000031
or a salt thereof.
3. The compound of claim 1, having the structure:
Figure FDA0003691104280000032
or a salt thereof.
4. The compound of claim 2, having the structure:
Figure FDA0003691104280000041
or a salt thereof.
5. A compound having the structure:
Figure FDA0003691104280000042
or a salt thereof.
6. A compound having the structure:
Figure FDA0003691104280000043
or a salt thereof.
7. A process for the preparation of a compound of formula A,
Figure FDA0003691104280000044
wherein
R A Is a fluorine compound, and the fluorine compound is,
R B is that
Figure FDA0003691104280000051
R C Is methyl, and
R D is a mixture of hydrogen and a nitrogen-containing compound,
the method comprises the following steps:
reacting a compound having the formula A-5 in the presence of a base
Figure FDA0003691104280000052
With a compound having the formula A-6
Figure FDA0003691104280000053
Contacting to provide a compound having formula a.
CN201910549014.1A 2014-01-24 2015-01-23 Diaryl macrocycles as modulators of protein kinases Active CN110317213B (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201461931506P 2014-01-24 2014-01-24
US61/931,506 2014-01-24
US201462049326P 2014-09-11 2014-09-11
US62/049,326 2014-09-11
US201562106301P 2015-01-22 2015-01-22
US62/106,301 2015-01-22
CN201580005705.5A CN106170289B (en) 2014-01-24 2015-01-23 The huge ring of the diaryl of regulator as protein kinase
PCT/US2015/012597 WO2015112806A2 (en) 2014-01-24 2015-01-23 Diaryl macrocycles as modulators of protein kinases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201580005705.5A Division CN106170289B (en) 2014-01-24 2015-01-23 The huge ring of the diaryl of regulator as protein kinase

Publications (2)

Publication Number Publication Date
CN110317213A CN110317213A (en) 2019-10-11
CN110317213B true CN110317213B (en) 2022-10-18

Family

ID=53682127

Family Applications (3)

Application Number Title Priority Date Filing Date
CN201580005705.5A Active CN106170289B (en) 2014-01-24 2015-01-23 The huge ring of the diaryl of regulator as protein kinase
CN201910549748.XA Active CN110317214B (en) 2014-01-24 2015-01-23 Diaryl macrocycles as modulators of protein kinases
CN201910549014.1A Active CN110317213B (en) 2014-01-24 2015-01-23 Diaryl macrocycles as modulators of protein kinases

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN201580005705.5A Active CN106170289B (en) 2014-01-24 2015-01-23 The huge ring of the diaryl of regulator as protein kinase
CN201910549748.XA Active CN110317214B (en) 2014-01-24 2015-01-23 Diaryl macrocycles as modulators of protein kinases

Country Status (31)

Country Link
US (5) US9714258B2 (en)
EP (6) EP3097107B1 (en)
JP (5) JP6490713B2 (en)
KR (1) KR102402179B1 (en)
CN (3) CN106170289B (en)
AP (1) AP2016009383A0 (en)
AU (2) AU2015209239B2 (en)
CA (1) CA2936079C (en)
CL (1) CL2016001876A1 (en)
CY (1) CY1121850T1 (en)
DK (2) DK3097107T3 (en)
EA (2) EA201892241A1 (en)
ES (2) ES2933350T3 (en)
HK (1) HK1231407A1 (en)
HR (2) HRP20221518T1 (en)
HU (2) HUE045208T2 (en)
IL (2) IL246860B (en)
LT (3) LT3636649T (en)
MX (2) MX2016009588A (en)
MY (1) MY193524A (en)
NZ (2) NZ761094A (en)
PE (1) PE20160931A1 (en)
PH (1) PH12016501463B1 (en)
PL (2) PL3097107T3 (en)
PT (2) PT3572416T (en)
RS (2) RS63829B1 (en)
SG (2) SG11201605929RA (en)
SI (2) SI3097107T1 (en)
UA (1) UA121206C2 (en)
WO (1) WO2015112806A2 (en)
ZA (2) ZA201604654B (en)

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS53350B (en) 2008-09-22 2014-10-31 Array Biopharma, Inc. Substituted imidazo[1,2b] pyridazine compounds as trk kinase inhibitors
EP3372605B1 (en) 2008-10-22 2021-11-03 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
AR077468A1 (en) 2009-07-09 2011-08-31 Array Biopharma Inc PIRAZOLO COMPOUNDS (1,5-A) PYRIMIDINE SUBSTITUTED AS TRK-QUINASA INHIBITORS
PL3205654T3 (en) 2010-05-20 2019-08-30 Array Biopharma, Inc. Macrocyclic compounds as trk kinase inhibitors
EA201892241A1 (en) 2014-01-24 2019-02-28 ТиПи ТЕРАПЬЮТИКС, ИНК. DIARYLIC MACRO CYCLES AS PROTEINKINASE MODULATORS
UA123044C2 (en) 2014-11-16 2021-02-10 Ерей Біофарма Інк. Crystalline form of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10758525B2 (en) 2015-01-22 2020-09-01 MyoKardia, Inc. 4-methylsulfonyl-substituted piperidine urea compounds
EP3722441A1 (en) 2015-06-01 2020-10-14 Loxo Oncology Inc. Method of diagnosing a cancer for a treatment with a trk inhibitor
ES2864839T3 (en) * 2015-07-02 2021-10-14 Turning Point Therapeutics Inc Chiral diaryl macrocycles as modulators of protein kinases
CN113354653A (en) * 2015-07-06 2021-09-07 特普医药公司 Diaryl macrocyclic polymorphs and methods of making the same
KR20180041135A (en) 2015-07-16 2018-04-23 어레이 바이오파마 인크. A pyrazolo [1,5-a] pyridine compound substituted as a RET kinase inhibitor
EP3325488B1 (en) * 2015-07-21 2020-06-24 Turning Point Therapeutics, Inc. Chiral diaryl macrocycle and use thereof in the treatment of cancer
EP3368039A1 (en) 2015-10-26 2018-09-05 The Regents of The University of Colorado, A Body Corporate Point mutations in trk inhibitor-resistant cancer and methods relating to the same
WO2017106505A1 (en) * 2015-12-15 2017-06-22 University Of South Florida Gas5 binding compounds, formulations, and uses thereof
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
BR112018070304A2 (en) 2016-04-04 2019-01-29 Loxo Oncology Inc (s) -n- (5 - ((r) -2- (2,5-difluorophenyl) -pyrrolidin-1-yl) -pyrazolo [1,5-a] pyrimidin-3-yl) -3 liquid formulations -hydroxypyrrolidine-1-carboxamide
TN2018000338A1 (en) 2016-04-04 2020-01-16 Loxo Oncology Inc Methods of treating pediatric cancers
BR112018073504A2 (en) 2016-05-18 2019-03-26 Array Biopharma, Inc. process for preparing (5) -n- (5 - ((r) -2- (2,5-difluorophenyl) pyrrolidin-1-yl) pyrazol [1,5-a] pyrimidin-3-yl) -3- hydroxypyrrolidine-1-carboxamide and salts thereof
JP2019527230A (en) * 2016-07-28 2019-09-26 ターニング・ポイント・セラピューティクス・インコーポレイテッドTurning Point Therapeutics,Inc. Macrocyclic kinase inhibitor
TWI704148B (en) 2016-10-10 2020-09-11 美商亞雷生物製藥股份有限公司 Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190077A1 (en) 2016-10-10 2019-04-09 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190092A1 (en) 2016-10-26 2019-04-25 Array Biopharma Inc PROCESS FOR THE PREPARATION OF PYRAZOLO[1,5-a]PYRIMIDINES AND SALTS THEREOF
CN110267960B (en) 2017-01-18 2022-04-26 阿雷生物药品公司 Substituted pyrazolo [1,5-a ] pyrazine compounds as RET kinase inhibitors
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
TWI808958B (en) * 2017-01-25 2023-07-21 美商特普醫葯公司 Combination therapy involving diaryl macrocyclic compounds
JOP20190213A1 (en) 2017-03-16 2019-09-16 Array Biopharma Inc Macrocyclic compounds as ros1 kinase inhibitors
CN107586796B (en) * 2017-07-20 2021-08-06 暨明医药科技(苏州)有限公司 Synthesis method of (R) -2- (1-aminoethyl) -4-fluorophenol
CA3069232A1 (en) 2017-07-28 2019-01-31 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
TWI791053B (en) 2017-10-10 2023-02-01 美商亞雷生物製藥股份有限公司 Crystalline forms of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile and pharmaceutical composition thereof
TWI812649B (en) 2017-10-10 2023-08-21 美商絡速藥業公司 Formulations of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
MA50456A (en) 2017-10-26 2020-09-02 Array Biopharma Inc MACROCYCLIC KINASE TRK INHIBITOR FORMULATIONS
CN109516999B (en) * 2017-11-01 2021-08-17 郑州泰基鸿诺医药股份有限公司 Compounds useful as protein kinase modulators and uses thereof
EP3706749B1 (en) 2017-11-10 2024-03-13 Angex Pharmaceutical, Inc. Macrocyclic compounds as trk kinase inhibitors and uses thereof
US20210087206A1 (en) 2017-12-19 2021-03-25 Turning Point Therapeutics, Inc. Macrocyclic kinase inhibitors and their use
PT3728271T (en) 2017-12-19 2022-10-06 Turning Point Therapeutics Inc Macrocyclic compounds for treating disease
CN111362949B (en) 2017-12-22 2021-12-21 深圳市塔吉瑞生物医药有限公司 Substituted pyrazolo [1,5-a ] pyrimidine compound, and pharmaceutical composition and application thereof
CA3087354C (en) 2018-01-18 2023-01-03 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
JP6997876B2 (en) 2018-01-18 2022-02-04 アレイ バイオファーマ インコーポレイテッド Substituted pyrazolyl [4,3-C] pyridine compound as a RET kinase inhibitor
JP7061195B2 (en) 2018-01-18 2022-04-27 アレイ バイオファーマ インコーポレイテッド Substituted pyrazolo [3,4-d] pyrimidine compound as a RET kinase inhibitor
EP3744723B1 (en) * 2018-01-23 2023-10-18 Shenzhen TargetRx, Inc. Substituted pyrazolo[1,5-a]pyrimidine macrocyclic compound
TW201932472A (en) * 2018-01-30 2019-08-16 大陸商上海吉倍生物技術有限公司 Compound having macrocyclic molecular structure and use thereof
CN110156813B (en) * 2018-02-13 2023-07-25 北京诺诚健华医药科技有限公司 Heterocyclic compounds as TRK inhibitors
JP2021519297A (en) * 2018-03-28 2021-08-10 フォチョン・ファーマシューティカルズ・リミテッドFochon Pharmaceuticals, Ltd. Macrocycle as a TRK kinase inhibitor
EP3773725A1 (en) 2018-03-29 2021-02-17 Loxo Oncology Inc. Treatment of trk-associated cancers
US11358973B2 (en) 2018-04-16 2022-06-14 Shenzhen Targetrx, Inc. Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity
CN111902417B (en) * 2018-04-25 2023-07-28 北京普祺医药科技股份有限公司 Diaryl macrocyclic compound, pharmaceutical composition and application thereof
CN111918868B (en) * 2018-05-04 2022-12-30 正大天晴药业集团股份有限公司 Diaryl macrocycles as protein kinase modulators
CN110627812B (en) 2018-06-25 2022-10-11 北京诺诚健华医药科技有限公司 Heterocyclic compounds as TRK inhibitors
CA3224949A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions, formulations, and polymorphs of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoropropan-2-yl)-1h-pyrazole-4-carboxamide
ES2922314T3 (en) 2018-09-10 2022-09-13 Array Biopharma Inc Fused Heterocyclic Compounds as RET Kinase Inhibitors
CN110950889B (en) * 2018-09-27 2022-04-05 北京赛林泰医药技术有限公司 Multi-target kinase inhibitor and preparation method and application thereof
WO2020063965A1 (en) * 2018-09-29 2020-04-02 南京明德新药研发有限公司 Pyrazolopyrimidine derivative as selective trk inhibitor
JP2022505987A (en) 2018-10-22 2022-01-14 エスカー セラピューティクス,インコーポレイテッド TYK2 inhibitors and their use
CN111171049B (en) * 2018-11-09 2021-06-04 山东轩竹医药科技有限公司 Tyrosine kinase inhibitors and uses thereof
SG11202104609SA (en) * 2018-11-09 2021-06-29 Shandong Xuanzhu Pharma Co Ltd Macrocyclic tyrosine kinase inhibitor and uses thereof
US20220081438A1 (en) 2018-12-19 2022-03-17 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
JP2022515197A (en) 2018-12-19 2022-02-17 アレイ バイオファーマ インコーポレイテッド 7-((3,5-dimethoxyphenyl) amino) quinoxaline derivative as an FGFR inhibitor for treating cancer
CN111592541B (en) * 2019-02-21 2021-09-10 山东轩竹医药科技有限公司 Macrocyclic kinase inhibitors and uses thereof
CN113727984B (en) * 2019-05-21 2024-03-15 浙江海正药业股份有限公司 Macrocyclic derivatives, preparation method and application thereof
WO2020257189A1 (en) * 2019-06-19 2020-12-24 Turning Point Therapeutics, Inc. Macrocycles for treating disease
PE20220135A1 (en) * 2019-06-19 2022-01-27 Turning Point Therapeutics Inc POLYMORPHS OF A MACROCYCLIC KINASE INHIBITOR
WO2020257165A1 (en) * 2019-06-19 2020-12-24 Turning Point Therapeutics, Inc. Macrocycles for use in treating disease
CN112110938B (en) * 2019-06-21 2021-11-09 成都海博为药业有限公司 Compound as protein kinase inhibitor and preparation method and application thereof
MA56505A (en) * 2019-06-21 2022-04-27 Janssen Pharmaceutica Nv MACROCYCLIC MCL-1 INHIBITORS
WO2021027503A1 (en) * 2019-08-12 2021-02-18 罗欣药业(上海)有限公司 Tricyclic compound, preparation method therefor, and intermediate and use thereof
WO2021063276A1 (en) * 2019-09-30 2021-04-08 浙江海正药业股份有限公司 Macrocyclic derivative, preparation method therefor and use thereof
WO2021108672A1 (en) 2019-11-27 2021-06-03 Turning Point Therapeutics, Inc. Combination therapy involving diaryl macrocyclic compounds
JP2023505056A (en) 2019-11-27 2023-02-08 ターニング・ポイント・セラピューティクス・インコーポレイテッド Combination therapy with diaryl macrocycles
PE20221485A1 (en) 2019-12-03 2022-09-26 Turning Point Therapeutics Inc MACROCYCLES FOR USE IN THE TREATMENT OF DISEASES
CN113045587A (en) * 2019-12-27 2021-06-29 成都倍特药业股份有限公司 Crystal form of macrocyclic compound and preparation method thereof
IL294214A (en) 2019-12-27 2022-08-01 Schr?Dinger Inc Cyclic compounds and methods of using same
CN113135938B (en) * 2020-01-19 2022-06-14 山东轩竹医药科技有限公司 Substituted macrocyclic tyrosine kinase inhibitors and uses thereof
CA3174455A1 (en) * 2020-03-02 2021-09-10 Turning Point Therapeutics, Inc. Therapeutic uses of macrocyclic compounds
WO2021223748A1 (en) * 2020-05-08 2021-11-11 山东轩竹医药科技有限公司 Crystal form of macrocyclic tyrosine kinase inhibitor and preparation method therefor
JP2023528907A (en) * 2020-06-04 2023-07-06 シンノハブ ファーマシューティカル カンパニー,リミティド Compound having macrocyclic structure and use thereof
EP4178958A1 (en) * 2020-07-10 2023-05-17 Blossomhill Therapeutics, Inc. Macrocycles and their use
US20230365584A1 (en) 2020-09-10 2023-11-16 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
EP4284804A1 (en) 2021-01-26 2023-12-06 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disorders
WO2022182845A1 (en) * 2021-02-25 2022-09-01 Blossomhill Therapeutics, Inc. Macrocycles and their use
TW202300150A (en) 2021-03-18 2023-01-01 美商薛定諤公司 Cyclic compounds and methods of using same
WO2023078267A1 (en) * 2021-11-02 2023-05-11 赛诺哈勃药业(成都)有限公司 Amino-containing macrocyclic compound as protein kinase modulator
WO2023133375A1 (en) * 2022-01-05 2023-07-13 Blossomhill Therapeutics, Inc. Macrocyclic compounds and use as kinase inhibitors
CN115746023A (en) * 2022-10-27 2023-03-07 复旦大学 Indazole structure-containing heterocyclic macrocyclic compound as protein kinase inhibitor and preparation method thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012136859A1 (en) * 2011-04-07 2012-10-11 Helmholtz-Zentrum für Infektionsforschung GmbH Medicament for liver regeneration and for treatment of liver failure
WO2013028465A1 (en) * 2011-08-19 2013-02-28 Merck Sharp & Dohme Corp. Crystal forms of a hcv protease inhibitor
CN102971322A (en) * 2010-05-20 2013-03-13 阵列生物制药公司 Macrocyclic compounds as TRK kinase inhibitors
CN104470523A (en) * 2012-03-09 2015-03-25 莱西肯医药有限公司 Imidazo [1, 2 - b] pyridazine - based compounds, compositions comprising them, and uses thereof

Family Cites Families (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4838925A (en) 1986-04-25 1989-06-13 E. I. Du Pont De Nemours And Company Heterocyclic acyl sulfonamides
EP0281841B1 (en) 1987-03-07 1990-11-14 Bayer Ag Process for preparation of 5-amino-4,6-dihalogenopyridines
WO1994021608A1 (en) 1993-03-25 1994-09-29 The Upjohn Company Indoletetralins having dopaminergic activity
FR2746309B1 (en) 1996-03-22 1998-04-17 Oreal COMPOSITION FOR DYEING KERATIN FIBERS CONTAINING PYRAZOLOPYRIMIDINEOXO; THEIR USE FOR DYEING AS COUPLER, DYEING PROCESSES
RS49779B (en) 1998-01-12 2008-06-05 Glaxo Group Limited, Byciclic heteroaromatic compounds as protein tyrosine kinase inhibitors
FR2793791B1 (en) 1999-05-19 2002-01-25 Univ Paris 7 Denis Diderot NOVEL INHIBITOR COMPOUNDS SPECIFIC TO A2 PHOSPHOLIPASES
DK1325008T3 (en) 2000-07-31 2006-02-13 Hoffmann La Roche piperazine
EP1379493A2 (en) 2001-03-23 2004-01-14 Eli Lilly and Company Non-imidazole aryl alkylamines compounds as histamine h3 receptor antagonists, preparation and therapeutic uses
US7550470B2 (en) 2002-12-11 2009-06-23 Merck & Co. Inc. Substituted pyrazolo[1,5-A]pyrimidines as tyrosine kinase inhibitors
EP2334301B1 (en) * 2008-09-08 2018-02-14 Merck Serono SA Macrocyclics pyrimidines as aurora kinase inhibitors
RS53350B (en) 2008-09-22 2014-10-31 Array Biopharma, Inc. Substituted imidazo[1,2b] pyridazine compounds as trk kinase inhibitors
EP3372605B1 (en) 2008-10-22 2021-11-03 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
AU2009308675A1 (en) 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
WO2010063487A1 (en) 2008-12-05 2010-06-10 Merz Pharma Gmbh & Co. Kgaa Pyrazolopyrimidines, a process for their preparation and their use as medicine
WO2010086040A1 (en) 2009-01-29 2010-08-05 Biomarin Iga, Ltd. Pyrazolo-pyrimidines for treatment of duchenne muscular dystrophy
AR077468A1 (en) 2009-07-09 2011-08-31 Array Biopharma Inc PIRAZOLO COMPOUNDS (1,5-A) PYRIMIDINE SUBSTITUTED AS TRK-QUINASA INHIBITORS
US20110029480A1 (en) * 2009-08-03 2011-02-03 IntelliCubes, Inc. Method of Compiling Multiple Data Sources into One Dataset
US9150577B2 (en) 2009-12-07 2015-10-06 Boehringer Ingelheim International Gmbh Heterocyclic compounds containing an indole core
EP2509979B1 (en) 2009-12-07 2016-05-25 Boehringer Ingelheim International GmbH Heterocyclic compounds containing a pyrrolopyridine or benzimidazole core
SA111320200B1 (en) 2010-02-17 2014-02-16 ديبيوفارم اس ايه Bicyclic Compounds and their Uses as Dual C-SRC / JAK Inhibitors
WO2012034095A1 (en) 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
US8637516B2 (en) 2010-09-09 2014-01-28 Irm Llc Compounds and compositions as TRK inhibitors
WO2012075393A2 (en) 2010-12-02 2012-06-07 President And Fellows Of Harvard College Activators of proteasomal degradation and uses thereof
WO2013001310A1 (en) * 2011-06-30 2013-01-03 Centro Nacional De Investigaciones Oncológicas (Cnio) Macrocyclic compounds and their use as cdk8 inhibitors
WO2013045702A2 (en) 2011-09-29 2013-04-04 L'oreal Dye composition comprising a non-glycosyl iridoid compound and a specific nucleophile or an amino or thio polymer, dyeing process, and device therefor
WO2013045701A2 (en) 2011-09-29 2013-04-04 L'oreal Dyeing process using a composition comprising a glycosyl iridoid compound and a nucleophile or an amino or thio polymer, composition and devices therefor
WO2013045653A1 (en) * 2011-09-30 2013-04-04 Oncodesign S.A. Macrocyclic flt3 kinase inhibitors
EP2768509B1 (en) 2011-10-20 2017-03-22 Glaxosmithkline LLC Substituted bicyclic aza-heterocycles and analogues as sirtuin modulators
AU2013229173B2 (en) 2012-03-06 2017-06-01 Pfizer Inc. Macrocyclic derivatives for the treatment of proliferative diseases
ES2676224T3 (en) 2012-03-09 2018-07-17 Lexicon Pharmaceuticals, Inc. Pyrazolo [1,5-a] pyrimidine-based compounds, compositions comprising them and methods for using them
GB201205669D0 (en) 2012-03-30 2012-05-16 Agency Science Tech & Res Bicyclic heterocyclic derivatives as mnk2 and mnk2 modulators and uses thereof
CA2899399A1 (en) 2013-01-30 2014-08-07 Bayer Pharma Aktiengesellschaft Amino-substituted isothiazoles
WO2014173928A1 (en) 2013-04-23 2014-10-30 Lek Pharmaceuticals D.D. Novel synthetic process to 8-chloro-1-methyl-benzo[d]azepine, novel intermediates and the production thereof
WO2015012328A1 (en) 2013-07-24 2015-01-29 武田薬品工業株式会社 Heterocyclic compound
WO2015073267A1 (en) 2013-11-15 2015-05-21 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
EA201892241A1 (en) 2014-01-24 2019-02-28 ТиПи ТЕРАПЬЮТИКС, ИНК. DIARYLIC MACRO CYCLES AS PROTEINKINASE MODULATORS
WO2016070241A1 (en) 2014-11-03 2016-05-12 Ctxt Pty Ltd Triazine compounds, compositions and synthesis
WO2016097869A1 (en) 2014-12-15 2016-06-23 Cmg Pharmaceutical Co., Ltd. Fused ring heteroaryl compounds and their use as trk inhibitors
EP3259264B1 (en) 2015-02-20 2022-07-27 Rigel Pharmaceuticals, Inc. Gdf-8 inhibitors
WO2016144844A1 (en) 2015-03-12 2016-09-15 Merck Sharp & Dohme Corp. Carboxamide inhibitors of irak4 activity
WO2016144846A1 (en) 2015-03-12 2016-09-15 Merck Sharp & Dohme Corp. Pyrazolopyrimidine inhibitors of irak4 activity
JP2018520992A (en) 2015-05-05 2018-08-02 バイエル ファーマ アクチエンゲゼルシャフト Cyclohexane derivatives substituted with amides
ES2864839T3 (en) 2015-07-02 2021-10-14 Turning Point Therapeutics Inc Chiral diaryl macrocycles as modulators of protein kinases
CN113354653A (en) 2015-07-06 2021-09-07 特普医药公司 Diaryl macrocyclic polymorphs and methods of making the same
EP3325488B1 (en) 2015-07-21 2020-06-24 Turning Point Therapeutics, Inc. Chiral diaryl macrocycle and use thereof in the treatment of cancer
EP3368039A1 (en) 2015-10-26 2018-09-05 The Regents of The University of Colorado, A Body Corporate Point mutations in trk inhibitor-resistant cancer and methods relating to the same
CN109153643B (en) 2016-03-04 2022-06-21 范德比尔特大学 Substituted indole MCL-1 inhibitors
JP2019527230A (en) 2016-07-28 2019-09-26 ターニング・ポイント・セラピューティクス・インコーポレイテッドTurning Point Therapeutics,Inc. Macrocyclic kinase inhibitor
EP3555137A1 (en) 2016-12-14 2019-10-23 Development Center for Biotechnology Antibody-drug conjugates and uses thereof
TWI808958B (en) 2017-01-25 2023-07-21 美商特普醫葯公司 Combination therapy involving diaryl macrocyclic compounds
CA3069232A1 (en) 2017-07-28 2019-01-31 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102971322A (en) * 2010-05-20 2013-03-13 阵列生物制药公司 Macrocyclic compounds as TRK kinase inhibitors
WO2012136859A1 (en) * 2011-04-07 2012-10-11 Helmholtz-Zentrum für Infektionsforschung GmbH Medicament for liver regeneration and for treatment of liver failure
WO2013028465A1 (en) * 2011-08-19 2013-02-28 Merck Sharp & Dohme Corp. Crystal forms of a hcv protease inhibitor
CN104470523A (en) * 2012-03-09 2015-03-25 莱西肯医药有限公司 Imidazo [1, 2 - b] pyridazine - based compounds, compositions comprising them, and uses thereof

Also Published As

Publication number Publication date
SG11201605929RA (en) 2016-08-30
JP2017503867A (en) 2017-02-02
MX2020001360A (en) 2020-03-20
HUE045208T2 (en) 2019-12-30
US20170002023A1 (en) 2017-01-05
CY1121850T1 (en) 2020-07-31
US20190169207A1 (en) 2019-06-06
CA2936079A1 (en) 2015-07-30
NZ761094A (en) 2023-02-24
CN106170289A (en) 2016-11-30
WO2015112806A2 (en) 2015-07-30
AP2016009383A0 (en) 2016-08-31
LT3572416T (en) 2022-12-27
KR20160111395A (en) 2016-09-26
ES2735729T3 (en) 2019-12-20
EP3636649A1 (en) 2020-04-15
EP3636650A1 (en) 2020-04-15
AU2015209239A1 (en) 2016-07-14
US9714258B2 (en) 2017-07-25
KR102402179B1 (en) 2022-05-25
PT3097107T (en) 2019-07-19
EP3572416B1 (en) 2022-09-21
SG10202000191YA (en) 2020-03-30
EP3097107B1 (en) 2019-04-17
US20200216465A1 (en) 2020-07-09
PL3572416T3 (en) 2023-02-27
LT3097107T (en) 2019-07-25
EP3097107A2 (en) 2016-11-30
EA201691492A1 (en) 2017-06-30
JP2021063121A (en) 2021-04-22
IL246860A0 (en) 2016-08-31
NZ722375A (en) 2022-11-25
US20220112213A1 (en) 2022-04-14
AU2019279951B2 (en) 2021-08-12
DK3097107T3 (en) 2019-07-08
EA031863B1 (en) 2019-03-29
HRP20191283T1 (en) 2019-10-18
EA201892241A1 (en) 2019-02-28
CA2936079C (en) 2022-07-19
CN106170289B (en) 2019-07-23
ZA201604654B (en) 2020-10-28
MY193524A (en) 2022-10-18
JP2023179539A (en) 2023-12-19
JP6490713B2 (en) 2019-03-27
JP2022133462A (en) 2022-09-13
MX2016009588A (en) 2017-05-15
IL246860B (en) 2019-09-26
JP7356546B2 (en) 2023-10-04
EP4338739A2 (en) 2024-03-20
HRP20221518T1 (en) 2023-02-17
AU2019279951A1 (en) 2020-01-16
CN110317214A (en) 2019-10-11
AU2015209239B2 (en) 2019-09-12
CN110317213A (en) 2019-10-11
UA121206C2 (en) 2020-04-27
PH12016501463A1 (en) 2017-02-06
RS63829B1 (en) 2023-01-31
CN110317214B (en) 2022-11-22
WO2015112806A3 (en) 2015-11-12
CL2016001876A1 (en) 2017-02-03
US10246466B2 (en) 2019-04-02
LT3636649T (en) 2024-04-10
PT3572416T (en) 2022-12-06
ES2933350T3 (en) 2023-02-06
ZA202004100B (en) 2023-12-20
HK1231407A1 (en) 2017-12-22
PL3097107T3 (en) 2020-01-31
PH12016501463B1 (en) 2017-02-06
IL268820B (en) 2021-10-31
RS59059B1 (en) 2019-08-30
EP3097107A4 (en) 2017-08-09
JP2019104748A (en) 2019-06-27
SI3097107T1 (en) 2019-08-30
IL268820A (en) 2019-10-31
EP3636648A1 (en) 2020-04-15
EP3636649B1 (en) 2024-02-14
PE20160931A1 (en) 2016-09-21
EP3572416A1 (en) 2019-11-27
DK3572416T3 (en) 2022-12-12
US10618912B2 (en) 2020-04-14
HUE060554T2 (en) 2023-03-28
US20170334929A1 (en) 2017-11-23
SI3572416T1 (en) 2023-02-28

Similar Documents

Publication Publication Date Title
CN110317213B (en) Diaryl macrocycles as modulators of protein kinases
CN107735399B (en) Chiral diaryl macrocycles as modulators of protein kinases
TWI688567B (en) Diaryl macrocycles as modulators of protein kinases
BR112016017101B1 (en) DIARYL MACROCYCLES COMPOUNDS AS MODULATORS OF PROTEIN KINASES AND THEIR PHARMACEUTICAL COMPOSITION
EA041662B1 (en) METHOD FOR PROTEIN-OR TYROSINE KINASE INHIBITION IN CELL USING DIARYL MACROCYCLIC COMPOUND

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40015633

Country of ref document: HK

GR01 Patent grant
GR01 Patent grant